[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (990)

Search Parameters:
Keywords = angiotensin II

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
35 pages, 3685 KiB  
Review
Molecular Basis of Na, K–ATPase Regulation of Diseases: Hormone and FXYD2 Interactions
by Bárbara Martins Cordeiro, Carlos Frederico Leite Fontes and José Roberto Meyer-Fernandes
Int. J. Mol. Sci. 2024, 25(24), 13398; https://doi.org/10.3390/ijms252413398 - 13 Dec 2024
Viewed by 407
Abstract
The Na, K–ATPase generates an asymmetric ion gradient that supports multiple cellular functions, including the control of cellular volume, neuronal excitability, secondary ionic transport, and the movement of molecules like amino acids and glucose. The intracellular and extracellular levels of Na+ and [...] Read more.
The Na, K–ATPase generates an asymmetric ion gradient that supports multiple cellular functions, including the control of cellular volume, neuronal excitability, secondary ionic transport, and the movement of molecules like amino acids and glucose. The intracellular and extracellular levels of Na+ and K+ ions are the classical local regulators of the enzyme’s activity. Additionally, the regulation of Na, K–ATPase is a complex process that occurs at multiple levels, encompassing its total cellular content, subcellular distribution, and intrinsic activity. In this context, the enzyme serves as a regulatory target for hormones, either through direct actions or via signaling cascades triggered by hormone receptors. Notably, FXYDs small transmembrane proteins regulators of Na, K–ATPase serve as intermediaries linking hormonal signaling to enzymatic regulation at various levels. Specifically, members of the FXYD family, particularly FXYD1 and FXYD2, are that undergo phosphorylation by kinases activated through hormone receptor signaling, which subsequently influences their modulation of Na, K–ATPase activity. This review describes the effects of FXYD2, cardiotonic steroid signaling, and hormones such as angiotensin II, dopamine, insulin, and catecholamines on the regulation of Na, K–ATPase. Furthermore, this review highlights the implications of Na, K–ATPase in diseases such as hypertension, renal hypomagnesemia, and cancer. Full article
(This article belongs to the Special Issue The Na, K-ATPase in Health and Disease)
Show Figures

Figure 1

Figure 1
<p><b>Crystal structure of the Na, K–ATPase in the E1.Mg<sup>2+</sup> state</b>. Panels (<b>A</b>,<b>B</b>) show the Na, K–ATPase from pig kidney viewed from two orthogonal directions. The enzyme consists of a catalytic α-subunit (orange), a glycosylated β-subunit (maroon), and a regulatory FXYD protein, specifically FXYD2 (yellow), located behind the α-subunit. The α-subunit comprises three well-defined cytoplasmic domains (A-blue, N-red, and P-gray) and 10 transmembrane helices (M1–M10). The helices are not labeled with numbers. Panel (<b>C</b>) highlights residues (pink) in the M9 region of the α subunit that interact with the corresponding FXYD peptides and are important for forming a stable complex [<a href="#B13-ijms-25-13398" class="html-bibr">13</a>]. Panel (<b>D</b>) shows the RRNS motif of the Na, K–ATPase α1 isoform, where Ser936 is phosphorylated by PKA [<a href="#B13-ijms-25-13398" class="html-bibr">13</a>]. Protein Data Bank (PDB) ID: 8JBL.</p>
Full article ">Figure 2
<p><b>Scheme of catalytic cycle of Na, K–ATPase</b>. Panel (<b>A</b>,<b>B</b>) represent the simplified Post–Albers model [<a href="#B11-ijms-25-13398" class="html-bibr">11</a>] that outlines the process of ATP hydrolysis and ion transport by the Na, K–ATPase. This enzyme alternates between two conformations, E1 and E2. In the forward cycle (clockwise), the Na, K–ATPase first binds intracellular Na<sup>+</sup> and MgATP with high affinity, forming the Na3E1ATP complex (Mg ions are not shown). The γ-phosphate of ATP is then transferred to the enzyme, and Na<sup>+</sup> ions become occluded (represented by parentheses). The resulting (Na3)E1-P<sup>•</sup>ADP complex has a high-energy phosphate bond, making the reaction reversible. After ADP is released, Na<sup>+</sup> ions are deoccluded and expelled into the extracellular space following or alongside the enzyme’s conformational shift in the enzyme to E2-P. This E2-P conformation also serves as the binding site for OUA, a well-known inhibitor of Na, K–ATPase. Extracellular K<sup>+</sup> then binds to E2-P, promoting Pi release and K<sup>+</sup> occlusion as they travel to the cytosol (protons, which are thought to bind to the “third Na<sup>+</sup> site” with two K<sup>+</sup> ions, are not shown). ATP, acting with low apparent affinity, accelerates K<sup>+</sup> deocclusion and intracellular release. The enzyme then shifts back from E2 to E1 and is ready to begin the cycle again.</p>
Full article ">Figure 2 Cont.
<p><b>Scheme of catalytic cycle of Na, K–ATPase</b>. Panel (<b>A</b>,<b>B</b>) represent the simplified Post–Albers model [<a href="#B11-ijms-25-13398" class="html-bibr">11</a>] that outlines the process of ATP hydrolysis and ion transport by the Na, K–ATPase. This enzyme alternates between two conformations, E1 and E2. In the forward cycle (clockwise), the Na, K–ATPase first binds intracellular Na<sup>+</sup> and MgATP with high affinity, forming the Na3E1ATP complex (Mg ions are not shown). The γ-phosphate of ATP is then transferred to the enzyme, and Na<sup>+</sup> ions become occluded (represented by parentheses). The resulting (Na3)E1-P<sup>•</sup>ADP complex has a high-energy phosphate bond, making the reaction reversible. After ADP is released, Na<sup>+</sup> ions are deoccluded and expelled into the extracellular space following or alongside the enzyme’s conformational shift in the enzyme to E2-P. This E2-P conformation also serves as the binding site for OUA, a well-known inhibitor of Na, K–ATPase. Extracellular K<sup>+</sup> then binds to E2-P, promoting Pi release and K<sup>+</sup> occlusion as they travel to the cytosol (protons, which are thought to bind to the “third Na<sup>+</sup> site” with two K<sup>+</sup> ions, are not shown). ATP, acting with low apparent affinity, accelerates K<sup>+</sup> deocclusion and intracellular release. The enzyme then shifts back from E2 to E1 and is ready to begin the cycle again.</p>
Full article ">Figure 3
<p><b>Representation of the chemical structures of hormones that regulate Na, K–ATPase</b>. The structures are represented as follows: Angiotensin II in green, dopamine in pink, epinephrine in yellow, norepinephrine in salmon, thyroxine in purple, and the three-dimensional structure of insulin (PDB: 1WAV). Oxygen atoms are shown in red, nitrogen atoms in blue, and iodine-123 in purple. The small molecule structures were obtained from ChemSpider, with CSIDs 150504, 661, 5611, 388394, and 64880242, respectively.</p>
Full article ">Figure 4
<p><b>Ang II induces multiple signaling pathways that regulate Na, K–ATPase activity</b>. In the adenylate cyclase–cAMP–PKA pathway, the phosphorylation of the α subunit by PKA inhibits Na, K–ATPase activity. Stimulatory effect of Ang II via the AT1R. The GRK4 increased phosphorylation of AT2R is associated with the inhibition of Na, K–ATPase. A signaling pathway involving PKC and the interaction of Na, K–ATPase with the adaptor protein 1 (AP1) recruits Na, K–ATPase molecules to the plasma membrane.</p>
Full article ">Figure 5
<p><b>Schematic diagram of the role of Na, K–ATPase in cell adhesion in different cancer types</b>. Blue line shows that Na<sup>+</sup> pump inhibition by CTS reduces cell adhesion in renal cells, leading to decreased expression and enzymatic activity, which is linked to cancer progression. Pink line shows that the overexpression β<sub>2</sub> isoform increased cellular adhesion on glia and ovary, arresting cancer progression.</p>
Full article ">Figure 6
<p><b>Signaling pathways of Na, K–ATPase and their consequences in cancer</b>. The inhibition of Na, K–ATPase by ouabain (red boxes) and the combined treatment with digoxin and cisplatin (green boxes) activate the Src/EGFR pathway, which leads to the activation of ERK and increased ROS production in mitochondria. This activates NF-κB, resulting in transcriptional regulation. EGFR phosphorylated activates the PKC pathway. Ouabain decreases the expression of alpha and beta subunits, while the combined treatment with digoxin and cisplatin has antitumoral effects. Solid arrows indicate experimentally supported events induced by inhibitors mentioned, while broken arrows indicate events with limited or indirect support.</p>
Full article ">
17 pages, 1649 KiB  
Review
COVID-19 Pathophysiology: Inflammation to Cardiac Injury
by Sami Fouda, Robert Hammond, Peter D Donnelly, Anthony R M Coates and Alexander Liu
Hearts 2024, 5(4), 628-644; https://doi.org/10.3390/hearts5040048 - 13 Dec 2024
Viewed by 1722
Abstract
Coronavirus disease 19 (COVID-19) is responsible for one of the worst pandemics in human history. The causative virus, the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), can invade host cells in multiple organs by binding the angiotensin-converting enzyme (ACE) II expressed on the [...] Read more.
Coronavirus disease 19 (COVID-19) is responsible for one of the worst pandemics in human history. The causative virus, the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), can invade host cells in multiple organs by binding the angiotensin-converting enzyme (ACE) II expressed on the cell surface. Once inside the host cell, viral replication takes place, leading to cellular disruption and the release of signal molecules that are recognised by the innate immune system. Innate immunity activation leads to the release of proinflammatory cytokines and primes the adaptive immune system. The proinflammatory environment defends against further viral entry and replication. SARS-CoV-2 infection is thought to lead to myocardial injury through several mechanisms. Firstly, direct viral-mediated cellular invasion of cardiomyocytes has been shown in in vitro and histological studies, which is related to cellular injury. Secondly, the proinflammatory state during COVID-19 can lead to myocardial injury and the release of protein remnants of the cardiac contractile machinery. Thirdly, the hypercoagulable state of COVID-19 is associated with thromboembolism of coronary arteries and/or other vascular systems. COVID-19 patients can also develop heart failure; however, the underlying mechanism is much less well-characterised than for myocardial injury. Several questions remain regarding COVID-19-related heart failure, including its potential reversibility, the role of anti-viral medications in its prevention, and the mechanisms underlying heart failure pathogenesis in long COVID-19. Further work is required to improve our understanding of the mechanism of cardiac sequelae in COVID-19, which may enable us to target SARS-CoV-2 and protect patients against longer-lasting cardiovascular complications. Full article
Show Figures

Figure 1

Figure 1
<p>Host cell infection by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Angiotensin-converting enzyme (ACE).</p>
Full article ">Figure 2
<p>Inflammatory activation in response to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infections. PAMPs: Pathogen Associated Molecular Patterns; DAMPs: Damage Associated Molecular Patterns.</p>
Full article ">Figure 3
<p>Adaptive immune system activation in Severe Acute Respiratory Syndrome Coronavirus 2 infections. MHC: Major Histocompatibility Complex.</p>
Full article ">Figure 4
<p>Potential mechanisms of myocardial injury in coronavirus disease 19.</p>
Full article ">Figure 5
<p>Central Illustration: BNP: B-type natriuretic peptide; CMR: cardiovascular magnetic resonance; ECG: electrocardiogram; SARS-CoV-2: severe acute respiratory syndrome coronavirus 2.</p>
Full article ">
16 pages, 1132 KiB  
Review
Angiotensin-(1-7) and Central Control of Cardiometabolic Outcomes: Implications for Obesity Hypertension
by Victoria L. Vernail, Lillia Lucas, Amanda J. Miller and Amy C. Arnold
Int. J. Mol. Sci. 2024, 25(24), 13320; https://doi.org/10.3390/ijms252413320 - 12 Dec 2024
Viewed by 238
Abstract
Hypertension is a leading independent risk factor for the development of cardiovascular disease, the leading cause of death globally. Importantly, the prevalence of hypertension is positively correlated with obesity, with obesity-related hypertension being difficult to treat due to a lack of current guidelines [...] Read more.
Hypertension is a leading independent risk factor for the development of cardiovascular disease, the leading cause of death globally. Importantly, the prevalence of hypertension is positively correlated with obesity, with obesity-related hypertension being difficult to treat due to a lack of current guidelines in this population as well as limited efficacy and adverse off-target effects of currently available antihypertensive therapeutics. This highlights the need to better understand the mechanisms linking hypertension with obesity to develop optimal therapeutic approaches. In this regard, the renin–angiotensin system, which is dysregulated in both hypertension and obesity, is a prime therapeutic target. While research and therapies have typically focused on the deleterious angiotensin II axis of the renin–angiotensin system, emerging evidence shows that targeting the protective angiotensin-(1-7) axis also improves cardiovascular and metabolic functions in animal models of obesity hypertension. While the precise mechanisms involved remain under investigation, in addition to peripheral actions, evidence exists to support a role for the central nervous system in the beneficial cardiometabolic effects of angiotensin-(1-7). This review will highlight emerging translational studies exploring the cardiovascular and metabolic regulatory actions of angiotensin-(1-7), with an emphasis on its central actions in brain regions including the brainstem and hypothalamus. An improved understanding of the central mechanisms engaged by angiotensin-(1-7) to regulate cardiovascular and metabolic functions may provide insight into the potential of targeting this hormone as a novel therapeutic approach for obesity-related hypertension. Full article
(This article belongs to the Special Issue Renin-Angiotensin System in Health and Diseases)
Show Figures

Figure 1

Figure 1
<p>RAS hormones elicit differential cardiometabolic effects by binding to specific angiotensin receptors. AT1R: angiotensin II type 1 receptor; AT2R: angiotensin II type 2 receptor; MasR: angiotensin-(1-7) Mas receptor; MrgD: Mas-related G protein-coupled receptor; SNS: sympathetic nervous system; PSNS: parasympathetic nervous system; BP: blood pressure; BRS: baroreflex sensitivity; NO: nitric oxide. Created in BioRender. Arnold, A. (2024) <a href="https://BioRender.com/e19f735" target="_blank">https://BioRender.com/e19f735</a> (accessed on 11 December 2024).</p>
Full article ">Figure 2
<p>Cross sections of cardiometabolic nuclei in the hypothalamus and brainstem of the mouse brain and their major physiological functions. PVN: the paraventricular nucleus of the hypothalamus; ARC: the arcuate nucleus of the hypothalamus; NTS: the nucleus tractus solitarius; RVLM: the rostral ventrolateral medulla; CVLM: the caudal ventrolateral medulla. The areas highlighted in red represent the circumventricular organs. OVLT: the organum vasculosum of the lamina terminalis; ME: the median eminence; SFO: the subfornical organ; AP: the area postrema. Dotted lines represent the approximate location of the cross sections displayed. Created in BioRender. Arnold, A. (2024) <a href="https://BioRender.com/o23u712" target="_blank">https://BioRender.com/o23u712</a> (accessed on 13 November 2024).</p>
Full article ">
22 pages, 7069 KiB  
Article
APOL1 Modulates Renin–Angiotensin System
by Vinod Kumar, Prabhjot Kaur, Kameshwar Ayasolla, Alok Jha, Amen Wiqas, Himanshu Vashistha, Moin A. Saleem, Waldemar Popik, Ashwani Malhotra, Christoph A. Gebeshuber, Karl Skorecki and Pravin C. Singhal
Biomolecules 2024, 14(12), 1575; https://doi.org/10.3390/biom14121575 - 10 Dec 2024
Viewed by 566
Abstract
Patients carrying APOL1 risk alleles (G1 and G2) have a higher risk of developing Focal Segmental Glomerulosclerosis (FSGS); we hypothesized that escalated levels of miR193a contribute to kidney injury by activating renin–angiotensin system (RAS) in the APOL1 milieus. Differentiated podocytes (DPDs) stably expressing [...] Read more.
Patients carrying APOL1 risk alleles (G1 and G2) have a higher risk of developing Focal Segmental Glomerulosclerosis (FSGS); we hypothesized that escalated levels of miR193a contribute to kidney injury by activating renin–angiotensin system (RAS) in the APOL1 milieus. Differentiated podocytes (DPDs) stably expressing vector (V/DPD), G0 (G0/DPDs), G1 (G1/DPDs), and G2 (G2/DPDs) were evaluated for renin, Vitamin D receptor (VDR), and podocyte molecular markers (PDMMs, including WT1, Podocalyxin, Nephrin, and Cluster of Differentiation [CD]2 associated protein [AP]). G0/DPDs displayed attenuated renin but an enhanced expression of VDR and Wilms Tumor [WT]1, including other PDMMs; in contrast, G1/DPDs and G2/DPDs exhibited enhanced expression of renin but decreased expression of VDR and WT1, as well as other PDMMs (at both the protein and mRNA levels). G1/DPDs and G2/DPDs also showed increased mRNA expression for Angiotensinogen and Angiotensin II Type 1 (AT1R) and 2 (AT2R) receptors. Protein concentrations of Brain Acid-Soluble Protein [BASP]1, Enhancer of Zeste Homolog [EZH]2, Histone Deacetylase [HDAC]1, and Histone 3 Lysine27 trimethylated [H3K27me3] in WT1-IP (immunoprecipitated proteins with WT1 antibody) fractions were significantly higher in G0/DPDs vs. G1/DPD and G2/DPDs. Moreover, DPD-silenced BASP1 displayed an increased expression of renin. Notably, VDR agonist-treated DPDs showed escalated levels of VDR and a higher expression of PDMMs, but an attenuated expression of renin. Human Embryonic Kidney (HEK) cells transfected with increasing APOL1(G0) plasmid concentrations showed a corresponding reduction in renin mRNA expression. Bioinformatics studies predicted the miR193a target sites in the VDR 3′UTR (untranslated region), and the luciferase assay confirmed the predicted sites. As expected, podocytes transfected with miR193a plasmid displayed a reduced VDR and an enhanced expression of renin. Renal cortical section immunolabeling in miR193a transgenic (Tr) mice showed renin-expressing podocytes. Kidney tissue extracts from miR193aTr mice also showed reduced expression of VDR and PDMMs, but enhanced expression of Renin. Blood Ang II levels were higher in miR193aTr, APOLG1, and APOL1G1/G2 mice when compared to control mice. Based on these findings, miR193a regulates the activation of RAS and podocyte molecular markers through modulation of VDR and WT1 in the APOL1 milieu. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

Figure 1
<p>Effect of APOL1 risk and non-risk alleles on podocyte expression of VDR, renin, and PDMMs. (<b>A</b>) Podocytes expressing Vector, G0, G1, and G2 were differentiated for 10 days. Differentiated podocytes (DPD) were harvested, and proteins were extracted and probed for renin and Podocalyxin (PDX) and reprobed for GAPDH (<span class="html-italic">n</span> = 4). Representative gels from two different lysates are shown. (<b>B</b>) Cumulative densitometric data (<span class="html-italic">n</span> = 4) are shown in bar graphs. Renin: * <span class="html-italic">p</span> &lt; 0.05 vs. V/DPD; # <span class="html-italic">p</span> &lt; 0.05 vs. G2/DPD; ## <span class="html-italic">p</span> &lt; 0.01 vs. G0/DPD.PDX: ** <span class="html-italic">p</span> &lt; 0.01 vs. VD/DPD; # <span class="html-italic">p</span> &lt; 0.05 vs. G0/DPD. (<b>C</b>) The lysates mentioned above (<b>A</b>) were also probed for Nephrin, VDR, CD2AP, WT1, and GAPDH (<span class="html-italic">n</span> = 4). Representative gels from two different lysates are displayed. (<b>D</b>) Cumulative densitometric data (<span class="html-italic">n</span> = 4) are shown in a bar diagram. Nephrin: * <span class="html-italic">p</span> &lt; 0.05 vs. V/DPD, G1/DPD, and G2/DPD; # <span class="html-italic">p</span> &lt; 0.05 vs. V/DPD. VDR: ** <span class="html-italic">p</span> &lt; 0.01 vs. V/DPD, G1/DPD, and G2/DPD; * <span class="html-italic">p</span> &lt; 0.05 vs. V/DPD; # <span class="html-italic">p</span> &lt; 0.05 vs. V/DPD; ## <span class="html-italic">p</span> &lt; 0.01 vs. G0/DPD. CD2AP: ** <span class="html-italic">p</span> &lt; 0.01 vs. V/DPD; # <span class="html-italic">p</span> &lt; 0.05 vs. G0/DPD. WT1: * <span class="html-italic">p</span> &lt; 0.05 vs. V/DPD; ## &lt; 0.01 vs. G0/DPD.</p>
Full article ">Figure 2
<p>Podocyte mRNA level alterations in APOL1 milieus. To determine mRNA levels of APOL1-expressing podocytes, RNAs were extracted from cellular lysates V/DPDs, G0/DPDs, G1/DPDs, and G2/DPDs (<span class="html-italic">n</span> = 4). cDNAs were amplified using specific primers (CD2AP, WT1, renin, VDR, and PDX [podocalyxin]). CD2AP: * <span class="html-italic">p</span> &lt; 0.05 with respective V and ** <span class="html-italic">p</span> &lt; 0.01 with respective G1 and G2; WT1: * <span class="html-italic">p</span> &lt; 0.05 with respective V and ** <span class="html-italic">p</span> &lt; 0.01 with respective G1 and G2; Renin: * <span class="html-italic">p</span> &lt; 0.05 with respective G0 and ** <span class="html-italic">p</span> &lt; 0.01 with respective G1 and G2; Nephrin: * <span class="html-italic">p</span> &lt; 0.05 with respective V and G0; VDR: ** <span class="html-italic">p</span> &lt; 0.01 with respective V and *** <span class="html-italic">p</span> &lt; 0.001 with respective G1 and G2; PDX: * <span class="html-italic">p</span> &lt; 0.05 with respective V, G1, and G2.</p>
Full article ">Figure 3
<p>miR1 93a expression in podocytes expressing Vector (V), APOL1G0, APOL1G1, and APOL1G2. Extracted RNAs (<span class="html-italic">n</span> = 4) were assayed for miR193a. Results (means ± SD) are displayed in a bar diagram. * <span class="html-italic">p</span> &lt; 0.05 vs. V; ** <span class="html-italic">p</span> &lt; 0.01 vs. V and G0.</p>
Full article ">Figure 4
<p>Renin-angiotensin status in APOL1 milieus. RNAs were extracted from V/DPD, G0/DPD, G1/DPD, and G2/DPD (<span class="html-italic">n</span> = 4), and cDNAs were amplified with specific primers for APOL1, renin, angiotensinogen, ACE1, AT1R, and AT2R. (<b>A</b>) APOL1: ** <span class="html-italic">p</span> &lt; 0.01 compared to V. (<b>B</b>) Renin: * <span class="html-italic">p</span> &lt; 0.05 compared to V; ** <span class="html-italic">p</span> &lt; 0.01 compared to V and G0; # <span class="html-italic">p</span> &lt; 0.05 compared to V and G1. (<b>C</b>) # <span class="html-italic">p</span> &lt; 0.05 compared to V, G1, and G2; * <span class="html-italic">p</span> &lt; 0.05 compared to V. (<b>D</b>) ** <span class="html-italic">p</span> &lt; 0.01 compared to V, G0, and G1. (<b>E</b>) AT1R: * <span class="html-italic">p</span> &lt; 0.05 compared to V, G0, and G2; ** <span class="html-italic">p</span> &lt; 0.01 compared to V and G0. (<b>F</b>) AT2R: * <span class="html-italic">p</span> &lt; 0.05 compared to V and G0; ** <span class="html-italic">p</span> &lt; 0.01 compared to V, G0, and G1.</p>
Full article ">Figure 5
<p>The structural construct of the WT1-BASP1 repressor complex. (<b>A</b>) Homology modeling and docking studies suggested the binding of WT1-BASP1 repressor complex on the renin promoter. (<b>B</b>) A schematic diagram displays the formation of the WT1-BASP1 repressor complex at the renin promoter. WT1 recruits BASP1, EZH2, and HDAC1, inducing methylation at Lysine 27 residues at Histone (H) 3 tail.</p>
Full article ">Figure 6
<p>Analysis of input lysates of podocytes expressing APOL1 non-risk and risk alleles. (<b>A</b>) Proteins were extracted from the cellular lysates of V/DPDs, G0/DPs, G1/DPDs, and G2/CDPs (<span class="html-italic">n</span> = 3–4). Gels from three independent lysates are displayed. (<b>B</b>) Cumulative densitometric data of proteins displayed in 3A are shown in a bar diagram. APOL1: ** <span class="html-italic">p</span> &lt; 0.01 vs. G1/DPD and G2/DPD; *** <span class="html-italic">p</span> &lt; 0.001 vs. V/DPD; Renin: * <span class="html-italic">p</span> &lt; 0.05 vs. respective variables; WT1: ## <span class="html-italic">p</span> &lt; 0.01 vs. respective variables; BASP1: # <span class="html-italic">p</span> &lt; 0.05 vs. respective variables; EZH2: <sup>a</sup> <span class="html-italic">p</span> &lt; 0.01 vs. V/DPD and G0/DPD; <sup>b</sup> <span class="html-italic">p</span> &lt; 0.01 vs. V/DPD and G0/DPD; HDAC1: <sup>c</sup> <span class="html-italic">p</span> &lt; 0.01 vs. respective other variables; H3K27me<sup>3</sup>: <sup>d</sup> <span class="html-italic">p</span> &lt; 0.01 vs. V/DPD, G1/DPD, and G2/DPD.</p>
Full article ">Figure 7
<p>Analysis of WT1 antibody-bound proteins (output). (<b>A</b>). Cellular lysates from the protocol of <a href="#biomolecules-14-01575-f006" class="html-fig">Figure 6</a>A were immunoprecipitated (IP) with the WT1 antibody. Protein blots of WT1-IP fractions were probed for WT1, renin, EZH2, HDAC1, H3K27me<sup>3</sup>, and IgG (<span class="html-italic">n</span> = 3). Gels from 3 independent cellular lysates are displayed. (<b>B</b>). Cumulative densitometric data from blots of the <a href="#biomolecules-14-01575-f007" class="html-fig">Figure 7</a>A (<span class="html-italic">n</span> = 3). WT1: * <span class="html-italic">p</span> &lt; 0.05 vs. V; ** <span class="html-italic">p</span> &lt; 0.01 vs. G1 and G2. BASP1: ** <span class="html-italic">p</span> &lt; 0.01 vs. respective other variables. EZH2: * <span class="html-italic">p</span> &lt; 0.05 vs. V; ** <span class="html-italic">p</span> &lt; 0.01 vs. G1; *** <span class="html-italic">p</span> &lt; 0.001 vs. G2. HDAC1: * <span class="html-italic">p</span> &lt; 0.05 vs. other variables. H3K27: * <span class="html-italic">p</span> &lt; 0.05 vs. V; ** <span class="html-italic">p</span> &lt; 0.01 vs. G1 and G2.</p>
Full article ">Figure 8
<p>Effect of BASP1 silencing on the podocyte expression of renin. (<b>A</b>) Cellular lysates of control podocytes (C/DPD), scrambled siRNA- (SCR/DPD), and BASP1-SiRNA-transfected podocytes (SiRNA/DPD) were probed for renin and GAPDH (<span class="html-italic">n</span> = 3). Gels of three independent lysates are shown. (<b>B</b>) Cumulative densitometric data (<span class="html-italic">n</span> = 3) are shown in bar graphs. ** <span class="html-italic">p</span> &lt; 0.01 vs. C/DPD and SCR/DPD.</p>
Full article ">Figure 9
<p>Effect of VDR overexpression on renin and podocyte molecular markers (PDMMs). (<b>A</b>) DPDs were incubated in media containing either vehicle (DMSO) or VDA (EB 1089, 10 nM) for 48 h (<span class="html-italic">n</span> = 4). Proteins were extracted and probed for VDR, renin, and PDMMs (WT1, Nephrin, CD2AP, and Synaptopodin) and GAPDH. Gels from two different lysates from V/DPD (vehicle-treated) and VDR/DPD (VDA-treated) are shown. (<b>B</b>) Cumulative densitometric data for different variables are shown in bar graphs. * <span class="html-italic">p</span> &lt; 0.0.5 and ** <span class="html-italic">p</span> &lt; 0.01 vs. respective V/DPD.</p>
Full article ">Figure 10
<p>Dose-response effect of APOL1 induction on renin expression in HEK cells. HEK cells were transfected with either empty vector (control, HEK-Cnt) or APOL1 plasmid (HEK-APOL1) in different concentrations (25, 50, and 100 ng) for 48 h (<span class="html-italic">n</span> = 4). Cells were harvested, and proteins and RNAs were extracted. (<b>A</b>) cDNAs were amplified with specific primers of APOL1 and renin. Cumulative data are shown in a bar diagram. APOL1 expression: # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 vs. HEK-Cnt. Renin: * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. HEK control; <span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01 vs. HEK-APOL1, 25 ng. (<b>B</b>) Proteins were probed for APOL1, renin, and GAPDH. Representative gels are displayed in the upper panel. Cumulative densitometric data are shown in bar graphs. APOL1: # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 vs. HEK-Cnt. Renin: * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. HEK-Cnt; <span>$</span><span>$</span> <span class="html-italic">p</span> &lt; 0.01 vs. HEK-APOL1, 25 ng.</p>
Full article ">Figure 11
<p>Validation of miR193a putative binding sites on VDR. (<b>A</b>) Available online in silico analysis tools (microrna.org; mirdb.org and TargetScan). VDR was predicted as a potential target for miR193a-5p. Predicted binding sites are shown. (<b>B</b>) Podocytes were transiently co-transfected by using Lipofectamine 2000 with wild-type or control reporter 3′-UTR plasmids and miR-193a (pCMV-miR-193a) or negative miR (control, AM17110) in combination. After 48 h of co-transfection, the firefly luciferase activities were measured using the duo-luciferase HS assay. The relative luciferase activity was calculated by normalizing it to Renilla luciferase. The presented results are cumulative values of three independent experiments, each performed in triplicate. *** <span class="html-italic">p</span> &lt; 0.001 vs. other variables.</p>
Full article ">Figure 12
<p>Effect of miR193a on the expression of VDR, renin, and PDMMs. (<b>A</b>) Podocytes were transfected with empty vector or miR19a plasmid and differentiated (<span class="html-italic">n</span> = 4). Cellular lysates were probed for VDR, renin, PDMMs (WT1, PDX, APOL1), and GAPDH. Gels from two different lysates are shown. (<b>B</b>) Cumulative data are shown in bar graphs. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.0.01 vs. respective V/DPD.</p>
Full article ">Figure 13
<p>Renal tissue expression profile of VDR, renin, and PDMMs in control and miR193aTr mice. (<b>A</b>) Renal tissues were harvested from control (Balb/C, wild-type) and miR193aTr mice (<span class="html-italic">n</span> = 3). Proteins were probed for renin, PDMMs (WT1, CD2AP, PDX), and GAPDH. Gels from three different lysates are displayed. (<b>B</b>) Tissue lysates from the above mice were reprobed for VDR, Nephrin, and GAPDH (<span class="html-italic">n</span> = 3). Gels from different lysates are shown. (<b>C</b>) Cumulative densitometric data from gels are shown in panel A in bar graphs. ** <span class="html-italic">p</span> &lt; 0.01 vs. control/Balb C mice. (<b>D</b>) Cumulative densitometric data from gels displayed in Panel B are shown in a bar diagram. ** <span class="html-italic">p</span> &lt; 0.0.01 vs. control/Balb C mice.</p>
Full article ">Figure 14
<p>Renal histology and VDR/renin expression in control (BALB/C) and miR193aTr mice. (<b>A</b>) Representative glomeruli from a control and miR193aTr mice. Sclerosis is displayed by black arrows in a glomerulus from an miR193aTr mouse. (<b>B</b>) Glomeruli were co-labeled with VDR and renin antibodies. Nuclei were stained with DAPI. A representative glomerulus from a control mouse showed co-labeled (VDR and renin) podocytes (white arrows); podocytes predominantly displayed green fluorescence (VDR) and minimal red fluorescence (renin). A representative glomerulus from an miR193aTr mouse displayed both green (VDR) and red (renin) fluorescence in podocytes (white arrows). Parietal epithelial cells showed orange fluorescence (combination of predominant red and mild green fluorescence, indicated by yellow arrows) in glomeruli from both BALB/C and miR193aTr mice. C. Blue square is magnified to display co-labeling of VDR and renin in podocytes. Scale bar = 50 µM.</p>
Full article ">Figure 15
<p>Ang II levels were determined in plasma samples of control (BALB/C, <span class="html-italic">n</span> = 6 and FVBN, <span class="html-italic">n</span> = 9) and experimental (miR193aTR, <span class="html-italic">n</span> = 6; APOL1 G0, <span class="html-italic">n</span> = 10; APOL1 G1, <span class="html-italic">n</span> = 9; and APOL1G1/G2, <span class="html-italic">n</span> = 9) mice. Results (means ± SD) are shown in bar diagrams. (<b>A</b>) Plasma Ang II levels in control (BALB/C) and miR193aTr mice. (<b>B</b>) Plasma Ang II levels in control and APOL1 mice. * <span class="html-italic">p</span> &lt; 0.05 vs. FVBN; # <span class="html-italic">p</span> &lt; 0.05 vs. APOL1G1.</p>
Full article ">Figure 16
<p>A schematic diagram displaying the activation of the RAS contributing to glomerular sclerosis in APOL1 milieus.</p>
Full article ">
39 pages, 6873 KiB  
Review
Exploring Mycolactone—The Unique Causative Toxin of Buruli Ulcer: Biosynthetic, Synthetic Pathways, Biomarker for Diagnosis, and Therapeutic Potential
by Gideon Atinga Akolgo, Kingsley Bampoe Asiedu and Richard Kwamla Amewu
Toxins 2024, 16(12), 528; https://doi.org/10.3390/toxins16120528 - 6 Dec 2024
Viewed by 907
Abstract
Mycolactone is a complex macrolide toxin produced by Mycobacterium ulcerans, the causative agent of Buruli ulcer. The aim of this paper is to review the chemistry, biosynthetic, and synthetic pathways of mycolactone A/B to help develop an understanding of the mode of [...] Read more.
Mycolactone is a complex macrolide toxin produced by Mycobacterium ulcerans, the causative agent of Buruli ulcer. The aim of this paper is to review the chemistry, biosynthetic, and synthetic pathways of mycolactone A/B to help develop an understanding of the mode of action of these polyketides as well as their therapeutic potential. The synthetic work has largely been driven by the desire to afford researchers enough (≥100 mg) of the pure toxins for systematic biological studies toward understanding their very high biological activities. The review focuses on pioneering studies of Kishi which elaborate first-, second-, and third-generation approaches to the synthesis of mycolactones A/B. The three generations focused on the construction of the key intermediates required for the mycolactone synthesis. Synthesis of the first generation involves assignment of the relative and absolute stereochemistry of the mycolactones A and B. This was accomplished by employing a linear series of 17 chemical steps (1.3% overall yield) using the mycolactone core. The second generation significantly improved the first generation in three ways: (1) by optimizing the selection of protecting groups; (2) by removing needless protecting group adjustments; and (3) by enhancing the stereoselectivity and overall synthetic efficiency. Though the synthetic route to the mycolactone core was longer than the first generation, the overall yield was significantly higher (8.8%). The third-generation total synthesis was specifically aimed at an efficient, scalable, stereoselective, and shorter synthesis of mycolactone. The synthesis of the mycolactone core was achieved in 14 linear chemical steps with 19% overall yield. Furthermore, a modular synthetic approach where diverse analogues of mycolactone A/B were synthesized via a cascade of catalytic and/or asymmetric reactions as well as several Pd-catalyzed key steps coupled with hydroboration reactions were reviewed. In addition, the review discusses how mycolactone is employed in the diagnosis of Buruli ulcer with emphasis on detection methods of mass spectrometry, immunological assays, RNA aptamer techniques, and fluorescent-thin layer chromatography (f-TLC) methods as diagnostic tools. We examined studies of the structure–activity relationship (SAR) of various analogues of mycolactone. The paper highlights the multiple biological consequences associated with mycolactone such as skin ulceration, host immunomodulation, and analgesia. These effects are attributed to various proposed mechanisms of actions including Wiskott–Aldrich Syndrome protein (WASP)/neural Wiskott–Aldrich Syndrome protein (N-WASP) inhibition, Sec61 translocon inhibition, angiotensin II type 2 receptor (AT2R) inhibition, and inhibition of mTOR. The possible application of novel mycolactone analogues produced based on SAR investigations as therapeutic agents for the treatment of inflammatory disorders and inflammatory pain are discussed. Additionally, their therapeutic potential as anti-viral and anti-cancer agents have also been addressed. Full article
(This article belongs to the Section Mycotoxins)
Show Figures

Figure 1

Figure 1
<p>Clinical forms of Buruli ulcer. (<b>A</b>) nodule, (<b>B</b>) plaque, (<b>C</b>) oedema, (<b>D</b>) small ulcer [<a href="#B9-toxins-16-00528" class="html-bibr">9</a>].</p>
Full article ">Figure 2
<p>Global map showing the distribution of Buruli ulcer disease as of 2023. Data source: World Health Organization. Map production: Control of Neglected Tropical Diseases (NTD), WHO [<a href="#B17-toxins-16-00528" class="html-bibr">17</a>].</p>
Full article ">Figure 3
<p>Structures of mycolactones A/B [<a href="#B89-toxins-16-00528" class="html-bibr">89</a>], C [<a href="#B90-toxins-16-00528" class="html-bibr">90</a>], D [<a href="#B91-toxins-16-00528" class="html-bibr">91</a>], E [<a href="#B91-toxins-16-00528" class="html-bibr">91</a>,<a href="#B92-toxins-16-00528" class="html-bibr">92</a>,<a href="#B93-toxins-16-00528" class="html-bibr">93</a>], E [<a href="#B91-toxins-16-00528" class="html-bibr">91</a>,<a href="#B92-toxins-16-00528" class="html-bibr">92</a>,<a href="#B93-toxins-16-00528" class="html-bibr">93</a>], F [<a href="#B78-toxins-16-00528" class="html-bibr">78</a>,<a href="#B94-toxins-16-00528" class="html-bibr">94</a>], <span class="html-italic">dia</span>-F [<a href="#B95-toxins-16-00528" class="html-bibr">95</a>,<a href="#B96-toxins-16-00528" class="html-bibr">96</a>], G [<a href="#B97-toxins-16-00528" class="html-bibr">97</a>], S1 [<a href="#B98-toxins-16-00528" class="html-bibr">98</a>], and S2 [<a href="#B98-toxins-16-00528" class="html-bibr">98</a>]. Lactone ring highlighted in red, C-linked C12–C20 side chain highlighted in blue, and polyunsaturated fatty acid side chain is in black.</p>
Full article ">Figure 4
<p>Complete structure of mycolactone A/B showing the core cyclic lactone ring (C1–C11) and two polyketide-derived highly unsaturated acyl side chains comprising the upper ‘Northern’ chain (C12–C20) and the longer ‘Southern’ chain (C1′–C16′). Under suitable laboratory conditions and light, mycolactone exists as geometric isomers centered around the double bond at C4′ C5′ in a 3:2 ratio.</p>
Full article ">Figure 5
<p>Overview of domain and module organization of the mycolactone PKS genes (<b>a</b>) <span class="html-italic">MlsA1</span> and <span class="html-italic">MlsA2</span> from the mycolactone PKS, harbored by the plasmid pMUM001 from <span class="html-italic">M. ulcerans</span> Agy99 [<a href="#B111-toxins-16-00528" class="html-bibr">111</a>,<a href="#B114-toxins-16-00528" class="html-bibr">114</a>]; (<b>b</b>) subunits (MLSA1, MLSA2, and MLSB) of different domains are represented by color block [<a href="#B115-toxins-16-00528" class="html-bibr">115</a>].</p>
Full article ">Figure 6
<p>Synthetic strategy employed by Kishi for the total syntheses of mycolactone A/B.</p>
Full article ">Figure 7
<p>Kishi’s synthetic strategies for the synthesis of suitably protected pentaenoic acid.</p>
Full article ">Figure 8
<p>Mycolactone A/B and proposed key steps of its total synthesis.</p>
Full article ">Figure 9
<p>Structure of truncated and biotinylated derivative of mycolactone synthetic (PG-204) for the detection of mAbs.</p>
Full article ">Figure 10
<p>Schematic illustration of Kishi’s improved mycolactone TLC detection method exploiting derivatization with 2-napthylboronic acid as a fluorescence enhancer [<a href="#B167-toxins-16-00528" class="html-bibr">167</a>].</p>
Full article ">Figure 11
<p>Proposed molecular targets and mechanisms of action for mycolactone-mediated ulcerative, immunosuppressive, and analgesic properties [<a href="#B58-toxins-16-00528" class="html-bibr">58</a>].</p>
Full article ">Figure 12
<p>Structure of mycolactone A/B and its synthetic analogues <b>56a</b> and <b>56b</b> with extended polyketide southern side chain.</p>
Full article ">Figure 13
<p>Synthetic analogues by Altmann and Pluschke.</p>
Full article ">Figure 14
<p>Blanchard synthetic analogues.</p>
Full article ">Scheme 1
<p>Synthesis of the C1–C7 fragment. Reagents and conditions: (1) Z-butene, <span class="html-italic"><sup>t</sup></span>BuOK, <span class="html-italic">n</span>-BuLi, (+)-(Ipc)<sub>2</sub>BOMe, BF<sub>3</sub>•OEt<sub>2</sub>, THF, −78 °C then NaOH, H<sub>2</sub>O<sub>2</sub>, 1 h, 80%; (2) TBSCl, imidazole, DMF, 96%; (3) O<sub>3</sub>, CH<sub>2</sub>Cl<sub>2</sub>, −78 °C, PPh<sub>3</sub>; (4) NaBH<sub>4</sub>, EtOH, 82% (2 steps); (5) Ph<sub>3</sub>P, I<sub>2</sub>, CH<sub>2</sub>Cl<sub>2</sub>, 88%.</p>
Full article ">Scheme 2
<p>Synthesis of the C8–C13 fragment. Reagents and conditions: (1) m-CPBA, CH<sub>2</sub>Cl<sub>2</sub>, 0 to 20 °C, 80%; (2) propyne, THF, <span class="html-italic">n</span>-BuLi, BF<sub>3</sub>•OEt<sub>2</sub>, −78 °C, 94%; (3) TBAF, THF, 73%; (4) cyclopentanone, TsOH, benzene, 76%; (5) Cp<sub>2</sub>ZrHCl, THF, 50 °C, 1 h; (6) I<sub>2</sub>, THF, 62%.</p>
Full article ">Scheme 3
<p>Synthesis of C14–C20 fragment. Reagents and conditions: (1) TsOH, MeOH/THF, 79%; (2) cyclopentanone, TsOH, 83%; (3) O<sub>3</sub>, CH<sub>2</sub>Cl<sub>2</sub> then PPh<sub>3</sub>, −78 °C, 97%; (4) DAMP, <span class="html-italic"><sup>t</sup></span>BuOK, THF, −78 °C, 88%; (5) <span class="html-italic">n</span>-BuLi, MeI, −78 to 20 °C, 99%; (6) Cp<sub>2</sub>ZrHCl, THF, 50 °C then I<sub>2</sub>, THF, 79%.</p>
Full article ">Scheme 4
<p>Kishi’s first-generation approach toward the synthesis of the fully hydroxy-protected core structure of mycolactones [<a href="#B102-toxins-16-00528" class="html-bibr">102</a>]. Reagents and conditions: (a) <span class="html-italic">t</span>-BuLi (3 equiv), ZnCl<sub>2</sub>, Pd(Ph<sub>3</sub>P)<sub>4</sub>, THF, 60%; (b) 1. CH<sub>2</sub>Cl<sub>2</sub>/H<sub>2</sub>O/TFA, (8:2:0.5), 77%; 2. PivCl, pyr., 99%; 3. TESCl, imid., CH<sub>2</sub>Cl<sub>2</sub>, 91%; 4. DiBAl-H, CH<sub>2</sub>Cl<sub>2</sub>, −78 °C, 98%; 5. I<sub>2</sub>, Ph<sub>3</sub>P imidazole, Et<sub>2</sub>O-MeCN (3:1), 91%; (c) <span class="html-italic">t</span>-BuLi (3 equiv), ZnCl<sub>2</sub>, Pd(Ph<sub>3</sub>P)<sub>4</sub>, THF, 50%; (d) 1. HF·pyr./pyr./THF (1:1:4), THF, 72%; 2. TEMPO, NCS, Bu<sub>4</sub>NCl, CH<sub>2</sub>Cl<sub>2</sub>-pH 8.6 buffer (1:1), 95%; 3. NaClO<sub>2</sub>, NaH<sub>2</sub>PO<sub>4</sub>, m-(MeO)<sub>2</sub>-C<sub>6</sub>H<sub>4</sub>, DMSO-<span class="html-italic">t</span>-BuOH (1:1), 94%; (e) 1. Cl<sub>3</sub>C<sub>6</sub>H<sub>2</sub>COCl, <span class="html-italic">i</span>-Pr<sub>2</sub>NEt, PhH; DMAP, PhH, 70%; 2. CH<sub>2</sub>Cl<sub>2</sub>/H<sub>2</sub>O/TFA (8:2:0.5), 62%; 3. HF·pyr., MeCN, 77%; (f) 1,1-dimethoxycyclopentane—25, <span class="html-italic">p</span>-TsOH, benzene, 80%.</p>
Full article ">Scheme 5
<p>Kishi’s second-generation approach toward the synthesis of the core structure of mycolactone. Reagents and conditions: (a) Zn, Cu(OAc)<sub>2</sub>, Pd(PPh<sub>3</sub>)<sub>4</sub>, LiCl, NMP, 60 °C, 83%; (b) <b>1</b>. CH<sub>2</sub>Cl<sub>2</sub>/H<sub>2</sub>O/TFA (16:4:1), 90%; <b>2</b>. TIPSCl, imidazole, DMF, 100%; <b>3</b>. LiOH, THF/MeOH/H<sub>2</sub>O, (4:1:1), 81%; (c) Cl<sub>3</sub>C<sub>6</sub>H<sub>2</sub>COCl, <span class="html-italic">i</span>-Pr<sub>2</sub>NEt, benzene, then DMAP, benzene 96%; (d) <b>1</b>. HF•py-py-CH<sub>3</sub>CN, 90%; <b>2</b>. Ph<sub>3</sub>P, imidazole, I<sub>2</sub>, CH<sub>2</sub>Cl<sub>2</sub>, 98%; (e) Zn, Cu(OAc)<sub>2</sub>, Pd(PPh<sub>3</sub>)<sub>4</sub>, LiCl, NMP, 60 °C, 80%; (f) DDQ, CH<sub>2</sub>Cl<sub>2</sub>/H<sub>2</sub>O, 91%.</p>
Full article ">Scheme 6
<p>Assembly of the mycolactone core. Reagents and conditions: (a) Zn, Cu(OAc)<sub>2</sub>, Pd(PPh<sub>3</sub>)<sub>4</sub>, LiCl; (b) TFA, wet CH<sub>2</sub>Cl<sub>2</sub>; (c) 1. TIPSOTf, 2,6-lutidine; 2. LiOH; (d) Cl<sub>3</sub>C<sub>6</sub>H<sub>2</sub>COCl, <span class="html-italic">i</span>-Pr<sub>2</sub>NEt, DMAP (e) HF·pyr., pyr.; (f) Ph<sub>3</sub>P, I<sub>2</sub> imidazole; (g) Zn, Cu(OAc)<sub>2</sub>, Pd(PPh<sub>3</sub>)<sub>4</sub>, LiCl.</p>
Full article ">Scheme 7
<p>Synthetic route for a suitably protected pentaenoic acid 49. Reagents and conditions: (a) LDA, THF, −78 °C, rt, 1 h, 94%; (b) LiOH, THF/MeOH/H2O (4:1:1), rt, 18 h, 100%.</p>
Full article ">Scheme 8
<p>Synthesis of the C9′–C16′ <span class="html-italic">tris</span>-TBS aldehyde 47. Reagents and conditions: (a) NaH, (EtO)<sub>2</sub>P(O)CH<sub>2</sub>CO<sub>2</sub>Et, THF, rt, 1 h, 64%; (b) AD-mix-a, MeSO<sub>2</sub>NH<sub>2</sub>, t-BuOH/H<sub>2</sub>O (1:1), 40 h, 0 °C, 70%; (c) <b>1</b>. TBSOTf, 2,6-lutidine, CH<sub>2</sub>Cl<sub>2</sub>, 0 °C, 99%; <b>2</b>. DIBAL, CH<sub>2</sub>Cl<sub>2</sub>, 89%; <b>3</b>. SO<sub>3·</sub>py, <span class="html-italic">i</span>-Pr<sub>2</sub>NEt, CH<sub>2</sub>Cl<sub>2</sub>-DMSO (3:2); <b>4</b>. Ph<sub>3</sub>P=C(Me)CO<sub>2</sub>Et, toluene, 110 °C, 83% (2 steps); (d) DIBAL, CH<sub>2</sub>Cl<sub>2</sub>, −78 °C, 57%; (e) SO<sub>3</sub>·py, i-Pr<sub>2</sub>NEt, CH<sub>2</sub>Cl<sub>2</sub>-DMSO (3:2) 100%.</p>
Full article ">Scheme 9
<p>Synthesis of the C<b>1′</b>–C<b>8′</b> phosphonate 46. Reagents and conditions: (a) 1. TBSCl, imidazole, DMF; 2. O<sub>3</sub>, CH<sub>2</sub>Cl<sub>2</sub>, −78 °C, then Ph<sub>3</sub>P; 3. Ph<sub>3</sub>P=C(Me)CO<sub>2</sub>Et, CH<sub>2</sub>Cl<sub>2</sub>; 4. DIBAL, CH<sub>2</sub>Cl<sub>2</sub>, −78 °C, 25% (four steps); (b) 1. SO<sub>3</sub>·py, i-Pr<sub>2</sub>NEt, CH<sub>2</sub>Cl<sub>2</sub>/DMSO (3:2); 2. Ph<sub>3</sub>P=C(Me)CO<sub>2</sub>Et, benzene, 90 °C, 80% (2 steps); (c) 1. DIBAL, CH<sub>2</sub>Cl<sub>2</sub>, −78 °C; 2. SO<sub>3</sub>·py, i-Pr<sub>2</sub>NEt, CH<sub>2</sub>Cl<sub>2</sub>/DMSO (3:2); 3. Ph3P=C(Me)CO<sub>2</sub>Et, benzene, 90 °C, 89% (3 steps); (d) 1. TBAF, THF, 87%; 2. PBr<sub>3</sub>, Et<sub>2</sub>O, 77%; 3. (EtO)<sub>3</sub>P, 90 °C, 96%.</p>
Full article ">Scheme 10
<p>Completion of Kishi’s first-generation total synthesis of mycolactone A/B. Reagents and conditions: (a) Cl<sub>3</sub>C<sub>6</sub>H<sub>2</sub>COCl, <span class="html-italic">i</span>-Pr<sub>2</sub>NEt, DMAP, PhH, rt, 20 h, 90%; (b) <b>1</b>. TBAF, THF, rt, 1 h, 81%; <b>2</b>. THF/HOAc/H<sub>2</sub>O (2:2:1), rt, 10 h, 67% with one recycle.</p>
Full article ">
20 pages, 6644 KiB  
Article
Host–Guest Complexation of Olmesartan Medoxomil by Heptakis(2,6-di-O-methyl)-β-cyclodextrin: Compatibility Study with Excipients
by Dana Emilia Man, Ema-Teodora Nițu, Claudia Temereancă, Laura Sbârcea, Adriana Ledeți, Denisa Ivan, Amalia Ridichie, Minodora Andor, Alex-Robert Jîjie, Paul Barvinschi, Gerlinde Rusu, Renata-Maria Văruţ and Ionuț Ledeți
Pharmaceutics 2024, 16(12), 1557; https://doi.org/10.3390/pharmaceutics16121557 - 4 Dec 2024
Viewed by 501
Abstract
Background: Olmesartan medoxomil (OLM) is the prodrug of olmesartan, an angiotensin II type 1 receptor blocker that has antihypertensive and antioxidant activities and renal protective properties. It exhibits low water solubility, which leads to poor bioavailability and limits its clinical potential. To improve [...] Read more.
Background: Olmesartan medoxomil (OLM) is the prodrug of olmesartan, an angiotensin II type 1 receptor blocker that has antihypertensive and antioxidant activities and renal protective properties. It exhibits low water solubility, which leads to poor bioavailability and limits its clinical potential. To improve the solubility of OLM, a host–guest inclusion complex (IC) between heptakis(2,6-di-O-methyl)-β-cyclodextrin (DMβCD) and the drug substance was obtained. Along with active substances, excipients play a crucial role in the quality, safety, and efficacy of pharmaceutical formulations. Therefore, the compatibility of OLM/DMβCD IC with several pharmaceutical excipients was evaluated. Methods: IC was characterized in both solid and liquid states, employing thermoanalytical techniques, universal-attenuated total reflectance Fourier-transform infrared spectroscopy, powder X-ray diffractometry, UV spectroscopy, and saturation solubility studies. Compatibility studies were carried out using thermal and spectroscopic methods to assess potential physical and chemical interactions. Results: The 1:1 OLM:DMβCD stoichiometry ratio and the value of the apparent stability constant were determined by means of the phase solubility method that revealed an AL-type diagram. The binary system showed different physicochemical characteristics from those of the parent entities, supporting IC formation. The geometry of the IC was thoroughly investigated using molecular modeling. Compatibility studies revealed a lack of interaction between the IC and all studied excipients at ambient conditions and the thermally induced incompatibility of IC with magnesium stearate and α-lactose monohydrate. Conclusions: The results of this study emphasize that OLM/DMβCD IC stands out as a valuable candidate for future research in the development of new pharmaceutical formulations, in which precautions should be considered in choosing magnesium stearate and α-lactose monohydrate as excipients if the manufacture stage requires temperatures above 100 °C. Full article
Show Figures

Figure 1

Figure 1
<p>Chemical structures of OLM (<b>a</b>) and DMβCD (<b>b</b>).</p>
Full article ">Figure 2
<p>Phase solubility diagram of OLM with DMβCD in 0.1 M phosphate buffer, pH 7.4.</p>
Full article ">Figure 3
<p>OLM/DMβCD IC simulation for 1:1 molar ratio. Images (<b>a</b>,<b>b</b>) show the supramolecular entity from the secondary face of the DMβCD cavity. OLM is represented as sticks colored by element, and DMβCD is represented by red/green/white dots (<b>a</b>); OLM is shown as spheres colored by element, and DMβCD is shown as sticks in red/green/white (<b>b</b>). Polar/hydrophobic contacts between OLM and DMβCD, where OLM is represented as sticks colored by element, and DMβCD is represented as lines (<b>c</b>). H-bond surface interaction of OLM/DMβCD (<b>d</b>).</p>
Full article ">Figure 4
<p>TG/DTG/DSC thermoanalytical curves of OLM (<b>a</b>); DMβCD (<b>b</b>); OLM/DMβCD PM (<b>c</b>); and KP (<b>d</b>) in air atmosphere.</p>
Full article ">Figure 5
<p>FTIR spectra of OLM, DMβCD, OLM/DMβCD PM, and KP.</p>
Full article ">Figure 6
<p>Diffraction profiles of OLM, DMβCD, and OLM/DMβCD binary systems PM and KP.</p>
Full article ">Figure 7
<p>UV spectra of DMβCD 150.0 µg mL<sup>−1</sup> and OLM 27.0 µg mL<sup>−1</sup> in 0.1 M phosphate buffer, pH 7.4, at 25 °C.</p>
Full article ">Figure 8
<p>TG (<b>a</b>,<b>b</b>), DTG (<b>c</b>,<b>d</b>), and DSC (<b>e</b>,<b>f</b>) curves of OLM/DMβCD IC and its mixture with pharmaceutical excipients TA and STA (<b>a</b>,<b>c</b>,<b>e</b>), and Mg STR and LA (<b>b</b>,<b>d</b>,<b>f</b>) in synthetic air atmosphere.</p>
Full article ">Figure 9
<p>UATR-FTIR spectra of (<b>a</b>) OLM/DMβCD IC, TA, STA, and the physical mixture of IC with TA and STA; (<b>b</b>) OLM/DMβCD IC, MgSTR, LA, and the mixture of IC with MgSTR and LA, recorded at ambient temperature.</p>
Full article ">Figure 10
<p>PXRD diffraction patterns of (<b>a</b>) OLM/DMβCD IC, TA, and their corresponding physical mixtures—main image; OLM/DMβCD KP + TA with 2θ values of diffraction peaks corresponding to KP—inset image; and OLM/DMβCD KP, excipients, and their mixture. (<b>b</b>) STA. (<b>c</b>) MgSTR. (<b>d</b>) LA.</p>
Full article ">Figure 10 Cont.
<p>PXRD diffraction patterns of (<b>a</b>) OLM/DMβCD IC, TA, and their corresponding physical mixtures—main image; OLM/DMβCD KP + TA with 2θ values of diffraction peaks corresponding to KP—inset image; and OLM/DMβCD KP, excipients, and their mixture. (<b>b</b>) STA. (<b>c</b>) MgSTR. (<b>d</b>) LA.</p>
Full article ">
28 pages, 1999 KiB  
Review
Pathophysiology of Angiotensin II-Mediated Hypertension, Cardiac Hypertrophy, and Failure: A Perspective from Macrophages
by Kelly Carter, Eshan Shah, Jessica Waite, Dhruv Rana and Zhi-Qing Zhao
Cells 2024, 13(23), 2001; https://doi.org/10.3390/cells13232001 - 4 Dec 2024
Viewed by 476
Abstract
Heart failure is a complex syndrome characterized by cardiac hypertrophy, fibrosis, and diastolic/systolic dysfunction. These changes share many pathological features with significant inflammatory responses in the myocardium. Among the various regulatory systems that impact on these heterogeneous pathological processes, angiotensin II (Ang II)-activated [...] Read more.
Heart failure is a complex syndrome characterized by cardiac hypertrophy, fibrosis, and diastolic/systolic dysfunction. These changes share many pathological features with significant inflammatory responses in the myocardium. Among the various regulatory systems that impact on these heterogeneous pathological processes, angiotensin II (Ang II)-activated macrophages play a pivotal role in the induction of subcellular defects and cardiac adverse remodeling during the progression of heart failure. Ang II stimulates macrophages via its AT1 receptor to release oxygen-free radicals, cytokines, chemokines, and other inflammatory mediators in the myocardium, and upregulates the expression of integrin adhesion molecules on both monocytes and endothelial cells, leading to monocyte-endothelial cell-cell interactions. The transendothelial migration of monocyte-derived macrophages exerts significant biological effects on the proliferation of fibroblasts, deposition of extracellular matrix proteins, induction of perivascular/interstitial fibrosis, and development of hypertension, cardiac hypertrophy and heart failure. Inhibition of macrophage activation using Ang II AT1 receptor antagonist or depletion of macrophages from the peripheral circulation has shown significant inhibitory effects on Ang II-induced vascular and myocardial injury. The purpose of this review is to discuss the current understanding in Ang II-induced maladaptive cardiac remodeling and dysfunction, particularly focusing on molecular signaling pathways involved in macrophages-mediated hypertension, cardiac hypertrophy, fibrosis, and failure. In addition, the challenges remained in translating these findings to the treatment of heart failure patients are also addressed. Full article
(This article belongs to the Special Issue New Insights into Vascular Biology in Health and Disease)
Show Figures

Figure 1

Figure 1
<p>Origin and development of tissue-resident and monocyte-derived macrophages. During the prenatal period (<b>A</b>), the tissue-resident macrophages (Mac) are generated during embryonic development through self-renewal proliferation in the yolk sac and fetal liver from erythro-myeloid progenitors (EMPs) and pre-macrophages (pMac), which can differentiate into long-lived tissue-resident macrophages. Distinct macrophage subpopulations crosstalk with specialized tissue cells, support tissue function, and maintain homeostasis during steady-state adulthood. During the early postnatal stage (<b>B</b>), bone marrow-derived hematopoietic stem cells (HSCs) give rise to short-lived and long-lived macrophages through a series of sequential differentiation. Conventionally, native macrophages, stimulated by cytokine-like lipopolysaccharides (LPS), interferon (IFN), colony-stimulating factor (CSF), and tumor necrosis factor (TNF), are polarized into Ly6c<sup>high</sup>CCR2<sup>+</sup> macrophages and release pro-inflammatory cytokines such as TNF-α, IL-1β, IL-6, IL-8, IL-12, and pro-fibrotic mediators TGF-β, IL-10, platelet derived growth factor (PDGF), and amphiregulin (AREG), which drive cell infiltration and tissue regeneration/remodeling.</p>
Full article ">Figure 2
<p>The pathological mechanisms underlying monocyte extravasation with angiotensin II (Ang II) stimulation. Transendothelial migration of monocytes into the interstitium involves the sequential interaction of distinct receptors on the surface of monocytes and endothelial cells. Ang II via the AT1 receptor upregulates the level of P-selectin glycoprotein ligand-1 (PSGL-1) on monocytes to mediate initial contact (rolling) between circulating monocytes and the vascular endothelium via upregulating lectin-like molecules (L-selectin) on monocytes and (P-selectin, E-selectin) on the endothelium. Subsequent monocyte firm adhesion to the endothelium relies on binding of the intercellular adhesion molecule-1 (ICAM-1) and the vascular cell adhesion molecule-1 (VCAM-1) to their ligands, including macrophage antigen 1 (Mac-1; CD11b/CD18) and lymphocyte function-associated antigen 1 (LFA-1; CD11a/CD18). This is followed by transendothelial migration, which is facilitated by additional Ig-supergene family member expression, including endothelial platelet-endothelial cell adhesion molecule (PECAM-1) and vascular endothelial (VE)-cadherin. Monocyte-derived C-C chemokine receptor 2 (CCR2<sup>+</sup>) macrophages in the interstitium induce myocardial injury by releasing various chemokines and cytokines.</p>
Full article ">Figure 3
<p>Schematic overview of angiotensin II (Ang II)-induced perivascular fibrosis and hypertension. Moving from left to right, depicting that the infiltration of monocyte-derived CCR2<sup>+</sup> macrophages in the aortic wall is a hallmark of Ang II-mediated vascular injury. On endothelium (Endo), Ang II facilitates transmigration of CCL2-expressed CCR2<sup>+</sup> macrophages by stimulating inflammatory responses, adhesion molecules (ICAMs), nuclear factor kappa light-chain-enhancer of activated B cells (NF-κB), and monocyte chemoattractant protein-1 (MCP-1). In vascular smooth muscle cells (VSMC), the vessel wall degradation and remodeling by Ang II are largely mediated through the production of NADPH oxidase, Ca<sup>2+</sup> overload, and the activation of G-protein couple receptor (GPCR) signaling. In adventitia, Ang II facilitates the infiltration of CCR2<sup>+</sup> macrophages by vasa vasorum neovascularization and stimulates the interaction between adventitial and perivascular tissues. Macrophages largely participate in vessel wall degradation and remodeling through the production of pro-inflammatory cytokines such as TNF-α, IL-1β, IL-10, and TGF-β1. Proliferation of fibroblasts (Fibs) to alpha smooth muscle actin (αSMA)-expressing myofibroblasts through the secretion of TGF-β from CCR2<sup>+</sup> macrophages cause the release of collagen assembly-related extracellular matrix proteins including cartilage oligomeric matrix protein (Comp), connective tissue growth factor (CTGF), and collagens, leading to perivascular fibrosis and hypertension through this maladaptive vascular remodeling process.</p>
Full article ">Figure 4
<p>Diagrammatic representation of physiological and pathological hypertrophic responses in the heart. In general, hypertrophy results in an increase in cardiomyocyte size and tissue mass depending upon physiological and pathophysiological stimuli. In exercise- and pregnancy-induced reversible physiological hypertrophy, the heart undergoes an individual cardiomyocyte increase in width with normal left ventricle (LV) cardiac function. In the physiological condition, cardiac resident Ly6C<sup>low</sup>CCR2<sup>−</sup> macrophages from the embryo act as cellular chaperones for tissue homeostasis via insulin-like growth factor-1 (IGF-1), amphiregulin (AREG), and growth factors (GFs). Pathological hypertrophy can be classified into pressure or volume overload (e.g., concentric vs. eccentric hypertrophy). Development of concentric hypertrophy is associated with hypertension and aortic stenosis, characterized as wall and septal thickening, a loss of chamber area, tissue fibrosis, and LV diastolic dysfunction. Over time, this state can deteriorate into dilated and eccentric hypertrophy, expressed as wall thinning, chamber dilatation, and an increase in wall tension along with LV systolic dysfunction. Some other disease states, such as myocardial infarction and dilated cardiomyopathy (DCM), can lead directly to eccentric hypertrophy without a prior concentric remodeling phase. In Ang II/AT1R-mediated concentric and eccentric hypertrophy, monocyte-derived Ly6C<sup>high</sup> CCR2<sup>+</sup> macrophages from the bone marrow promote the proliferation of myofibroblasts and lead to cardiac tissue fibrosis by releasing cytokines and depositing collagens and fibronectins.</p>
Full article ">Figure 5
<p>Role of macrophages in Ang II and pressure overload induced adverse cardiac remodeling and failure. A multifaceted pathological stimulation increases Ang II level and pressure overload, results in maladaptive remodeling, and leads to heart failure with a preserved ejection fraction (HFpEF) and diastolic dysfunction or a reduced ejection fraction (HFrEF) and systolic dysfunction. In the adaptive stage or the remote zone after myocardial infarction (MI), the tissue C-C chemokine receptor (CCR2<sup>−</sup>) resident macrophages maintain cardiac homeostasis, proliferation, and angiogenesis, promote tissue repair and regeneration, and preserve cardiac function, whereas in the conditions of concentric hypertrophy mediated by Ang II stimulation or pressure overload, as well as eccentric hypertrophy after MI, the replacement of CCR2<sup>−</sup> resident macrophages by recruited CCR2<sup>+</sup> macrophages in the myocardium or infarct zone evoke inflammation, induce fibrosis and scar formation, and deteriorate cardiac function. Depending upon the alterations in the degree of cardiac remodeling, e.g., extracellular matrix deposition, heart failure with HFpEF may develop to the stage of HFrEF. Although there is a controversial issue regarding the treatment of patients with HFpEF using angiotensin-converting enzyme inhibitors (ACEIs) or angiotensin receptor blockers (ARBs), both drugs significantly inhibit tissue fibrosis, preserve cardiac function, and reduce mortality in patients with HFrEF.</p>
Full article ">
11 pages, 2987 KiB  
Article
Characteristics of Cerebrovascular Response to Intrinsic Vasoactive Substances in Sika Deer (Cervus nippon yesoensis) and the Possible Effects of Gravity on Adrenergic Responses
by Md. Zahorul Islam, Siyuan Wu, Tomoki Ootawa, Henry Smith, Ha Thi Thanh Nguyen, Etsumori Harada and Atsushi Miyamoto
Animals 2024, 14(23), 3500; https://doi.org/10.3390/ani14233500 - 4 Dec 2024
Viewed by 467
Abstract
Gravity may exert species-specific effects on quadrupedal vasoreactivity, reflecting variations in the vertical displacement of the cardiocranial axis from the dorsal plane. Deer show markedly displaced cardiocranial axes compared to their closest phylogenetic relatives, but their relative cerebrovascular responses remain unelucidated. Accordingly, we [...] Read more.
Gravity may exert species-specific effects on quadrupedal vasoreactivity, reflecting variations in the vertical displacement of the cardiocranial axis from the dorsal plane. Deer show markedly displaced cardiocranial axes compared to their closest phylogenetic relatives, but their relative cerebrovascular responses remain unelucidated. Accordingly, we investigated the responses to noradrenaline (NA), acetylcholine (ACh), 5-hydroxytryptamine (5-HT), histamine, angiotensin (Ang) II, and bradykinin (BK) in cervine basilar arterial rings. NA and 5-HT induced slight contraction, and ACh induced relaxation, which contrasts with the findings reported in pigs and cattle. The cumulative response to ACh was abolished by endothelial denudation and inhibited by Nω-nitro-L-arginine (a nitric oxide synthase inhibitor), atropine (a nonselective muscarinic antagonist), and p-fluoro-hexahydro-sila-difenidol (an M3 antagonist). Pirenzepine (an M1 antagonist) and methoctramine (an M2 antagonist) showed no significant effects. Histamine induced contractions, with its concentration–response curve shifted to the right in parallel by diphenhydramine (an H1 antagonist). However, cimetidine (an H2 antagonist) showed no significant effects. Ang II and BK had no vasomotive effects. NA and ACh induced different cerebrovascular responses in sika deer versus cattle, but histamine and BK did not. Our findings suggest that cerebrovascular responses are influenced by the similarity of animal species and the head and heart positions relative to gravity. Full article
(This article belongs to the Section Mammals)
Show Figures

Figure 1

Figure 1
<p>Main arteries of the cerebral base in deer. Dorsal view.</p>
Full article ">Figure 2
<p>Effect of histamine (○), acetylcholine (●), noradrenaline (△), 5−hydroxytryptamine (<tt>▲</tt>), angiotensin II (+), bradykinin (×), and on isolated basilar arteries of deer. Contraction response was measured under resting tension and calculated as percent response to 60 mmol/L KCl, and relaxation in response to acetylcholine was assessed in the arteries precontracted with U-46619 (10<sup>−7</sup> mol/L) and calculated as percent response to 10<sup>−4</sup> mol/L sodium nitroprusside. Each point represents mean ± SEM of five deer.</p>
Full article ">Figure 3
<p>The concentration–response curves of histamine in endothelium-intact (●) and denuded (○) basilar arteries and the effect of cimetidine (△; 10<sup>−6</sup> mol/L) on histamine-induced contraction (●) in the endothelium-intact basilar arteries of deer. Cimetidine had no effects on the vascular resting tension or the histamine-induced contraction. The contractions induced by 60 mmol/L KCl were considered 100%. Each point represents the mean ± SEM of five deer.</p>
Full article ">Figure 4
<p>Effects of diphenhydramine (○, 10<sup>−8</sup>; △, 10<sup>−7</sup>; □, 10<sup>−6</sup> mol/L) on histamine-induced contractions (●) (<b>a</b>) and the Schild plot for diphenhydramine (<b>b</b>) in the basilar artery of deer. Contractions induced by 10<sup>−3</sup> mol/L histamine in the absence of diphenhydramine were considered 100%. Diphenhydramine competitively inhibited histamine-induced contraction. Values are expressed as mean ± SEM of five deer. CR, the ratio of equally effective histamine concentrations [50% maximal concentration (EC<sub>50</sub>) in the presence of diphenhydramine/EC<sub>50</sub> in the absence of diphenhydramine].</p>
Full article ">Figure 5
<p>Concentration–response curves of acetylcholine in endothelium-intact (●) and denuded (○) basilar arteries and effect of L-NNA (△, 10<sup>−4</sup> mol/L) on acetylcholine-induced relaxation (●) (<b>a</b>). L-NNA inhibited acetylcholine-induced relaxation. Effect of atropine (○, 10<sup>−7</sup> mol/L; △, 10<sup>−5</sup> mol/L) on acetylcholine-induced relaxation (●) in isolated basilar artery of deer (<b>b</b>). Atropine competitively inhibited acetylcholine-induced relaxation. U-46619 (10<sup>−7</sup> mol/L) was used for precontraction. Relaxation induced by 10<sup>−4</sup> mol/L sodium nitroprusside was considered 100%. Each point represents mean ± SEM for five deer.</p>
Full article ">Figure 6
<p>Effect of pirenzepine (○, 10<sup>−6</sup> mol/L), methoctramine (△, 10<sup>−6</sup> mol/L) (<b>a</b>), and pFHHSiD (○: 10<sup>−7</sup> mol/L, △: 10<sup>−6</sup> mol/L) (<b>b</b>) on acetylcholine induced relaxation (●) and Schild plot of pFHHSiD (<b>c</b>) for basilar artery of deer. U-46619 (10<sup>−7</sup> mol/L) were used for precontraction. Relaxation induced by 10<sup>−4</sup> mol/L sodium nitroprusside was considered 100%. Each point represents mean ± SEM for five deer. CR, equally effective ratio of ACh concentrations (EC<sub>50</sub> in presence of pFHHSiD/EC<sub>50</sub> in absence of pFHHSiD).</p>
Full article ">Figure 7
<p>(<b>a</b>) Representative examples of the vertical displacement (angle) of the cardiocranial axis from the dorsal plane in even-toed ungulates (deer, cattle and pigs). (<b>b</b>) Relationship between the response to noradrenaline and the vertical displacement in mammalian species (dolphins, pigs, cattle, mice, horses, dogs, and deer, n = 3 per species). Data for animals other than dogs and cattle were obtained using the same experimental methods.</p>
Full article ">
23 pages, 7444 KiB  
Article
Monocyte/Macrophage-Specific Loss of ARNTL Suppresses Chronic Kidney Disease-Associated Cardiac Impairment
by Yuya Yoshida, Naoki Nishikawa, Kohei Fukuoka, Akito Tsuruta, Kaita Otsuki, Taiki Fukuda, Yuma Terada, Tomohito Tanihara, Taisei Kumamoto, Ryotaro Tsukamoto, Takumi Nishi, Kosuke Oyama, Kengo Hamamura, Kouta Mayanagi, Satoru Koyanagi, Shigehiro Ohdo and Naoya Matsunaga
Int. J. Mol. Sci. 2024, 25(23), 13009; https://doi.org/10.3390/ijms252313009 - 3 Dec 2024
Viewed by 428
Abstract
Defects in Aryl hydrocarbon receptor nuclear translocator-like 1 (ARNTL), a central component of the circadian clock mechanism, may promote or inhibit the induction of inflammation by monocytes/macrophages, with varying effects on different diseases. However, ARNTL’s role in monocytes/macrophages under chronic kidney disease (CKD), [...] Read more.
Defects in Aryl hydrocarbon receptor nuclear translocator-like 1 (ARNTL), a central component of the circadian clock mechanism, may promote or inhibit the induction of inflammation by monocytes/macrophages, with varying effects on different diseases. However, ARNTL’s role in monocytes/macrophages under chronic kidney disease (CKD), which presents with systemic inflammation, is unclear. Here, we report that the expression of Arntl in monocytes promoted CKD-induced cardiac damage. The expression of G-protein-coupled receptor 68 (GPR68), which exacerbates CKD-induced cardiac disease, was regulated by ARNTL. Under CKD conditions, GPR68 expression was elevated via ARNTL, particularly in the presence of PU.1, a transcription factor specific to monocytes and macrophages. In CKD mouse models lacking monocyte-specific ARNTL, GPR68 expression in monocytes was reduced, leading to decreased cardiac damage and fibrosis despite no improvement in renal excretory capacity or renal fibrosis and increased angiotensin II production. The loss of ARNTL did not affect the expression of marker molecules, indicating the origin or differentiation of cardiac macrophages, but affected GPR68 expression only in cardiac macrophages derived from mature monocytes, highlighting the significance of the interplay between GPR68 and ARNTL in monocytes/macrophages and its influence on cardiac pathology. Understanding this complex relationship between circadian clock mechanisms and disease could help uncover novel therapeutic strategies. Full article
(This article belongs to the Topic Molecular and Cellular Mechanisms of Heart Disease)
Show Figures

Figure 1

Figure 1
<p>Effect of ARNTL on induction of GPR68 expression in RAW264.7 and mouse primary monocytes by 5/6Nx-derived serum. (<b>A</b>) mRNA of <span class="html-italic">Arntl</span> in RAW264.7 incubated with 10% serum from Sham and 5/6Nx mice for 24 h. (<b>B</b>) Transcriptional regulation of <span class="html-italic">Gpr68</span> using serum prepared from Sham or 5/6Nx mice. Number of nucleotide residues indicates distance from transcription start site (+1). RAW264.7 cells were transfected with <span class="html-italic">Gpr68</span> (-1734)-Luc, <span class="html-italic">Gpr68</span> (-1512)-Luc, <span class="html-italic">Gpr68</span> (-1261)-Luc, <span class="html-italic">Gpr68</span> (-27)-Luc, or pGL4.18. Values are expressed as mean ± S.D. (<span class="html-italic">n</span> = 4). (<b>C</b>) Influence of CLOCK/ARNTL on transcriptional activity of mouse <span class="html-italic">GPR68</span>. RAW264.7 cells were transfected with <span class="html-italic">Gpr68</span> (-1734)-Luc in presence or absence of CLOCK and ARNTL-expressing vectors. Relative luciferase activity of pGL4.18-transfected cells in absence of CLOCK/ARNTL was set at 1.0. (<b>D</b>) High-ARNTL-expressing RAW264.7 was created by introducing an ARNTL expression plasmid. ARNTL expression levels were measured using Western blotting. (<b>E</b>) Protein levels of GPR68 in RAW264.7-transfected pcDNA3.1 or ARNTL-expressing vectors. (<b>F</b>) Loss of <span class="html-italic">Arntl</span> caused by CRE-LOXP system resulted in loss of ARNTL protein in monocytes. Monocytes isolated from monocytic <span class="html-italic">ARNTL</span> +/+ mice or monocytic <span class="html-italic">ARNTL</span> −/− mice. (<b>G</b>) Expression of <span class="html-italic">Gpr68</span> mRNA in primary cultured monocytes, which were isolated from monocytic <span class="html-italic">ARNTL</span> +/+ mice or monocytic <span class="html-italic">ARNTL</span> −/− mice. mRNA levels of <span class="html-italic">Gpr68</span> were assessed after treatment with serum from Sham or 5/6Nx WT mice for 24 h. Values are expressed as mean ± S.D. (<span class="html-italic">n</span> = 4–6). *, <span class="html-italic">p</span> &lt; 0.05, **, <span class="html-italic">p</span> &lt; 0.01 indicates significant differences between two groups (two-way ANOVA with Tukey–Kramer post hoc tests or Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 2
<p>The effect of monocyte/macrophage-specific transcription factor PU.1 on the induction of GPR68 expression. (<b>A</b>) The 5/6Nx-derived serum did not increase the transcriptional activity upstream of <span class="html-italic">Gpr68</span> in NIH3T3. NIH3T3 was transfected with <span class="html-italic">Gpr68</span> (-1734)-Luc or pGL4.18 and incubated with 10% serum from Sham and 5/6Nx mice for 24 h. (<b>B</b>) Transcription factors binding upstream of <span class="html-italic">Gpr68</span> analyzed by previous transcriptome analyses. The blue waveform shows the sequenced tags in ChIP sequence analysis for each transcription factor. The numbers on the horizontal axis indicate the distance from the transcription start site (kbp). (<b>C</b>) The PU.1 protein in RAW264.7 incubated with 10% serum from Sham and 5/6Nx mice for 24 h. (<b>D</b>) High-PU.1-expressing NIH3T3 was created by introducing a PU.1 expression plasmid. PU.1 expression levels were measured using Western blotting. (<b>E</b>,<b>F</b>) The mRNA levels of <span class="html-italic">Gpr68</span> (<b>E</b>) and <span class="html-italic">Arntl</span> (<b>F</b>) in NIH3T3-transfected pcDNA3.1 or PU.1-expressing vectors were measured after incubation with 10% serum from Sham and 5/6Nx mice for 24 h. (<b>G</b>) A schematic of mouse <span class="html-italic">Gpr68</span>. The numbers indicate the distance from the transcription start site (+1). Black rectangles, E-box. The arrow symbols indicate the location on the gene where the primer sets localize for the analysis of ChIP. (<b>H</b>) The binding of endogenous ARNTL to the <span class="html-italic">Gpr68</span> upstream region in NIH3T3-transfected pcDNA3.1 or PU.1-expressing vectors. Values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 3–5). **, <span class="html-italic">p</span> &lt; 0.01 indicates significant differences between the two groups (two-way ANOVA with Tukey–Kramer post hoc tests or Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 3
<p>The effect of the loss of monocyte-specific ARNTL on the 5/6Nx-induced induction of GPR68 expression. (<b>A</b>) The binding of endogenous ARNTL or CLOCK to the <span class="html-italic">Gpr68</span> upstream region in Ly6G<sup>−</sup>/CD11b<sup>+</sup>/Ly6C<sup>+</sup> cells prepared from <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice in the blood. The primer sets used are shown in <a href="#ijms-25-13009-f002" class="html-fig">Figure 2</a>G. (<b>B</b>) The expression of <span class="html-italic">Gpr68</span> mRNA in Ly6G<sup>−</sup>/CD11b<sup>+</sup>/Ly6C<sup>+</sup> cells prepared from <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice in the blood and spleen. (<b>C</b>,<b>D</b>) Flow cytometry analysis was performed to detect high-GPR68-expressing Ly6G<sup>−</sup>/CD11b<sup>+</sup>/Ly6C<sup>+</sup> cells in the blood and spleen. The ratio of high-GPR68-expressing monocytes in the blood and spleen. For all panels, values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 5–7). **, <span class="html-italic">p</span> &lt; 0.01, ** indicates significant differences between the two groups (two-way ANOVA with Tukey–Kramer post hoc tests).</p>
Full article ">Figure 4
<p>The effect of the deficiency of monocyte-specific ARNTL on 5/6Nx-induced cardiac injury. (<b>A</b>) Serum BNP concentrations in <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. Values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 6). (<b>B</b>,<b>C</b>) Mutations in <span class="html-italic">Arntl</span> in monocytes ameliorated CKD-induced cardiac fibrosis. Panel (B) shows Masson’s trichrome staining of tissue fibrosis in blue. Scale bars indicate 1 mm (upper panel) and 50 μm (lower panel). Panel (<b>C</b>) shows the quantification of the fibrosis area under light microscopy. Values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 5). (<b>D</b>) The total amount of collagen throughout the ventricle. Values were corrected for total protein mass. Values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 5–6). (<b>E</b>) Cardiac TIMP-1 protein levels in <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. Values were corrected for total protein mass. Values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 5–6). (<b>F</b>) The mRNA levels of <span class="html-italic">Tnf-α</span> and <span class="html-italic">Il-6</span> and fibrosis-related factors (<span class="html-italic">Col1a1</span>, <span class="html-italic">Col1a2</span>, <span class="html-italic">Mmp1a</span>, <span class="html-italic">Timp-1</span>, and <span class="html-italic">αSma</span>) in the cardiac ventricle of <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. The mean value of the Sham-operated <span class="html-italic">ARNTL</span> +/+ group was set to 1.0. Values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 5). *, <span class="html-italic">p</span> &lt; 0.05, **, <span class="html-italic">p</span> &lt; 0.01 indicates significant differences between the two groups (two-way ANOVA with Tukey–Kramer post hoc tests).</p>
Full article ">Figure 5
<p>The effect of the loss of monocyte-specific ARNTL on renal function in 5/6Nx mice. (<b>A</b>) Masson’s trichrome staining for the kidneys prepared from <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. Scale bars indicate 50 μm. (<b>B</b>) The total amount of collagen throughout the kidney. Values were corrected for total protein mass. Values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 5–6). (<b>C</b>) Renal TIMP-1 protein levels in <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. Values were corrected for total protein mass. Values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 5–6). (<b>D</b>) The mRNA levels of <span class="html-italic">Tnf-α</span> and <span class="html-italic">Il-6</span> and fibrosis-related factors (<span class="html-italic">Col1a1</span>, <span class="html-italic">Col1a2</span>, <span class="html-italic">Mmp1a</span>, <span class="html-italic">Timp-1</span>, and <span class="html-italic">αSma</span>) in the kidney of <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. The mean value of the Sham-operated <span class="html-italic">ARNTL</span> +/+ group was set to 1.0. Values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 5). (<b>E</b>–<b>H</b>) The serum concentrations of creatinine (<b>E</b>), urea nitrogen (<b>F</b>), angiotensin II (<b>G</b>), and aldosterone (<b>H</b>), in <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. (<b>I</b>) The mRNA levels of <span class="html-italic">Tgf-β</span> in the kidney of <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. The mean value of the Sham-operated <span class="html-italic">ARNTL</span> +/+ group was set as 1.0. (<b>J</b>) The serum concentrations of retinol in <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. In all panels, values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 4–5). **, <span class="html-italic">p</span> &lt; 0.01; *, <span class="html-italic">p</span> &lt; 0.05 significant difference between the two groups (one-way or two-way ANOVA with Tukey–Kramer post hoc tests).</p>
Full article ">Figure 6
<p>The effect of the loss of monocyte-specific ARNTL on the 5/6Nx-induced induction of GPR68 expression. (<b>A</b>,<b>B</b>) The mRNA levels of <span class="html-italic">Vcam1</span> and <span class="html-italic">Sele</span> in the cardiac ventricle or kidney of <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. The mean value of the Sham-operated <span class="html-italic">ARNTL</span> +/+ group was set to 1.0. (<b>C</b>,<b>D</b>) The number of cardiac or renal F4/80<sup>+</sup>/Ly6G<sup>−</sup>/CD11b<sup>+</sup>/Ly6C<sup>+</sup> cells (<b>C</b>) and F4/80<sup>+</sup>/Ly6G<sup>−</sup>/CD11b<sup>+</sup>/Ly6C<sup>−</sup> cells (<b>D</b>) in each organ. The mean value of the Sham-operated <span class="html-italic">ARNTL</span> +/+ group in each organ was set as 1.0. (<b>E</b>) The mRNA levels of <span class="html-italic">Gpr68</span> in the cardiac ventricle or kidney of <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. The mean value of the Sham-operated <span class="html-italic">ARNTL</span> +/+ group was set to 1.0. (<b>F</b>) The expression of <span class="html-italic">Gpr68</span> mRNA in cardiac F4/80<sup>+</sup>/Ly6G<sup>−</sup>/CD11b<sup>+</sup>/Ly6C<sup>+</sup> and F4/80<sup>+</sup>/Ly6G<sup>−</sup>/CD11b<sup>+</sup>/Ly6C<sup>−</sup> cells prepared from <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice ventricles. (<b>G</b>) The expression levels in the cardiac F4/80<sup>+</sup>/Ly6G<sup>−</sup>/CD11b<sup>+</sup>/Ly6C<sup>+</sup> and F4/80<sup>+</sup>/Ly6G<sup>−</sup>/CD11b<sup>+</sup>/Ly6C<sup>−</sup> cells of markers indicative of a subset of macrophages. Histograms showing the expression of each marker were obtained by flow cytometric analysis. For all panels, values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 4–7). *, <span class="html-italic">p</span> &lt; 0.05, **, <span class="html-italic">p</span> &lt; 0.01 indicates significant differences between the two groups (two-way ANOVA with Tukey–Kramer post hoc tests).</p>
Full article ">
10 pages, 647 KiB  
Review
Vasopressor Therapy
by Jean-Louis Vincent and Filippo Annoni
J. Clin. Med. 2024, 13(23), 7372; https://doi.org/10.3390/jcm13237372 - 3 Dec 2024
Viewed by 485
Abstract
Vasopressor therapy represents a key part of intensive care patient management, used to increase and maintain vascular tone and thus adequate tissue perfusion in patients with shock. Norepinephrine is the preferred first-line agent because of its reliable vasoconstrictor effects, with minimal impact on [...] Read more.
Vasopressor therapy represents a key part of intensive care patient management, used to increase and maintain vascular tone and thus adequate tissue perfusion in patients with shock. Norepinephrine is the preferred first-line agent because of its reliable vasoconstrictor effects, with minimal impact on heart rate, and its mild inotropic effects, helping to maintain cardiac output. Whichever vasopressor is used, its effects on blood flow must be considered and excessive vasoconstriction avoided. Other vasoactive agents include vasopressin, which may be considered in vasodilatory states, and angiotensin II, which may be beneficial in patients with high renin levels, although more data are required to confirm this. Dobutamine should be considered, along with continued fluid administration, to help maintain adequate tissue perfusion in patients with reduced oxygen delivery. In this narrative review, we consider the different vasopressor agents, focusing on the importance of tailoring therapy to the individual patient and their hemodynamic response. Full article
(This article belongs to the Section Emergency Medicine)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the principal effects of adrenergic agents on the alpha- and beta-adrenergic receptors.</p>
Full article ">Figure 2
<p>The four components of cardiac output and how it can be increased (arrows).</p>
Full article ">
13 pages, 2027 KiB  
Article
Antihypertensive Effect of Perla and Esmeralda Barley (Hordeum vulgare L.) Sprouts in an Induction Model with L-NAME In Vivo
by Abigail García-Castro, Alma D. Román-Gutiérrez, Fabiola A. Guzmán-Ortiz and Raquel Cariño-Cortés
Metabolites 2024, 14(12), 678; https://doi.org/10.3390/metabo14120678 - 3 Dec 2024
Viewed by 546
Abstract
Background: Hypertension is one of the leading causes of premature death worldwide. Despite advances in conventional treatments, there remains a significant need for more effective and natural alternatives to control hypertension. In this context, sprouted barley extracts have emerged as a potential therapeutic [...] Read more.
Background: Hypertension is one of the leading causes of premature death worldwide. Despite advances in conventional treatments, there remains a significant need for more effective and natural alternatives to control hypertension. In this context, sprouted barley extracts have emerged as a potential therapeutic option. This study presents the evaluation of the bioactive properties of extracts from two varieties of barley germinated for different periods (3, 5, and 7 days), focusing on their potential to regulate blood pressure mechanisms. Objectives/Methods: The main objective was to assess the effects of these extracts on blood pressure regulation in N(ω)-Nitro-L-Arginine Methyl Ester (L-NAME)-induced hypertensive rats. Renal (creatinine, urea, uric acid, and total protein) and endothelial (NOx levels) function, angiotensin-converting enzyme (ACE) I and II activity, and histopathological effects on heart and kidney tissues were evaluated. Results: In particular, Esmeralda barley extract demonstrated 83% inhibition of ACE activity in vitro. Furthermore, the combined administration of sprouted barley extract (SBE) and captopril significantly reduced blood pressure and ACE I and II activity by 22%, 81%, and 76%, respectively, after 3, 5, and 7 days of germination. The treatment also led to reductions in protein, creatinine, uric acid, and urea levels by 3%, 38%, 42%, and 48%, respectively, along with a 66% increase in plasma NO concentrations. Conclusions: This study highlights the bioactive properties of barley extracts with different germination times, emphasizing their potential health benefits as a more effective alternative to conventional antihypertensive therapies. Full article
(This article belongs to the Special Issue Plants and Plant-Based Foods for Metabolic Disease Prevention)
Show Figures

Figure 1

Figure 1
<p>Decrease in a ngiotensin-converting e nzyme activity (percent). Bars represent the mean ± SD. The significance levels are represented by the value of <span class="html-italic">p</span> &lt; 0.05 (* <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.0001) compared to the control group (c aptopril). Different letters indicate significant differences between varieties and the same day of germination. ANOVA followed by a Tukey test was performed.</p>
Full article ">Figure 2
<p>(<b>a</b>) Effects of barley extracts and captopril supplementation on serum ACE I activity. (<b>b</b>) Effects of barley extracts and captopril supplementation on kidney ACE II activity. Each bar represents mean ± SD. Significance levels indicated by <span class="html-italic">p</span> &lt; 0.05 (* <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.0001) when compared with L-NAME group (ANOVA followed by Tukey test).</p>
Full article ">Figure 3
<p>Histopathological changes in cardiac muscle (H-E stain). (<b>A</b>) Normotensive group, normal cardiac. The arrow indicates the transverse striation of the longitudinal section of the cardiac muscle.; (<b>B</b>) L-NAME group, chronic inflammation; (<b>C</b>) L-NAME + Captopril group, inflammatory cells decreased inflammation; (<b>D</b>) L-NAME + Esmeralda group, inflammatory cells are observed, moderate inflammation; (<b>E</b>) L-NAME + Perla group, reduction in inflammation; (<b>F</b>) L-NAME + Perla + Esmeralda group, without histopathological changes; (<b>G</b>) L-NAME Esmeralda + Captopril group, inflammation relief. The arrows in (<b>B</b>–<b>D</b>), indicate the infiltration of proinflammatory cells.</p>
Full article ">Figure 4
<p>Histopathological changes in renal glomerulus. The arrows indicate the following changes in the renal glomerulus: (<b>A</b>) Normotensive group, without observable changes; (<b>B</b>) L-NAME group, degeneration and necrosis of renal glomerulus; (<b>C</b>) L-NAME + Captopril group, partial adhesion of glomerulus to Bowman’s capsules; (<b>D</b>) L-NAME + Esmeralda group, slight glomerular necrosis; (<b>E</b>) L-NAME + Perla group, Bowman space dilation; (<b>F</b>) L-NAME + Perla + Esmeralda group, decreased inflammation of renal glomerulus; (<b>G</b>) L-NAME Esmeralda + Captopril group, without observable histological changes.</p>
Full article ">
18 pages, 1973 KiB  
Article
Angiotensin II Exposure In Vitro Reduces High Salt-Induced Reactive Oxygen Species Production and Modulates Cell Adhesion Molecules’ Expression in Human Aortic Endothelial Cell Line
by Nikolina Kolobarić, Nataša Kozina, Zrinka Mihaljević and Ines Drenjančević
Biomedicines 2024, 12(12), 2741; https://doi.org/10.3390/biomedicines12122741 - 29 Nov 2024
Viewed by 422
Abstract
Background/Objectives: Increased sodium chloride (NaCl) intake led to leukocyte activation and impaired vasodilatation via increased oxidative stress in human/animal models. Interestingly, subpressor doses of angiotensin II (AngII) restored endothelium-dependent vascular reactivity, which was impaired in a high-salt (HS) diet in animal models. [...] Read more.
Background/Objectives: Increased sodium chloride (NaCl) intake led to leukocyte activation and impaired vasodilatation via increased oxidative stress in human/animal models. Interestingly, subpressor doses of angiotensin II (AngII) restored endothelium-dependent vascular reactivity, which was impaired in a high-salt (HS) diet in animal models. Therefore, the present study aimed to assess the effects of AngII exposure following high salt (HS) loading on endothelial cells’ (ECs’) viability, activation, and reactive oxygen species (ROS) production. Methods: The fifth passage of human aortic endothelial cells (HAECs) was cultured for 24, 48, and 72 h with NaCl, namely, the control (270 mOsmol/kg), HS320 (320 mOsmol/kg), and HS350 (350 mOsmol/kg). AngII was administered at the half-time of the NaCl incubation (10−4–10−7 mol/L). Results: The cell viability was significantly reduced after 24 h in the HS350 group and in all groups after longer incubation. AngII partly preserved the viability in the HAECs with shorter exposure and lower concentrations of NaCl. Intracellular hydrogen peroxide (H2O2) and peroxynitrite (ONOO) significantly increased in the HS320 group following AngII exposure compared to the control, while it decreased in the HS350 group compared to the HS control. A significant decrease in superoxide anion (O2.−) formation was observed following AngII exposure at 10−5, 10−6, and 10−7 mol/L for both HS groups. There was a significant decrease in intracellular adhesion molecule 1 (ICAM-1) and endoglin expression in both groups following treatment with 10−4 and 10−5 mol/L of AngII. Conclusions: The results demonstrated that AngII significantly reduced ROS production at HS350 concentrations and modulated the viability, proliferation, and activation states in ECs. Full article
(This article belongs to the Special Issue Renin-Angiotensin System in Cardiovascular Biology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Cellular metabolic activity of HAECs after treatment with different concentrations of NaCl and AngII assessed via MTT assay. A—absorbance; nm—nanometers; CTRL—control; HS—high salt; AngII—angiotensin II; One-way ANOVA; *<sup>,+</sup> significance level <span class="html-italic">p</span> &lt; 0.05 (* compared to control group; <sup>+</sup> compared to HS group before AngII).</p>
Full article ">Figure 2
<p>Formation of hydrogen peroxide and peroxynitrite in HAECs following high-salt treatment accompanied by AngII exposure (<b>A</b>). Representative histogram overlay (<b>B</b>) and stacked histograms (<b>C</b>). Grey color representing CTRL group, blue color representing HS320 group, red color representing HS350 group. Results are expressed as geometric mean fluorescence intensity (GMFI). DCF-DA—dichlorofluorescein diacetate; CTRL—control; HS—high salt; One-way ANOVA; *<sup>,+</sup> significance level <span class="html-italic">p</span> &lt; 0.05 (* compared to HS group before AngII; <sup>+</sup> compared to control group).</p>
Full article ">Figure 3
<p>Formation of superoxide in HAECs following high-salt treatment accompanied by AngII exposure (<b>A</b>). Representative histogram overlay (<b>B</b>) and stacked histograms (<b>C</b>). Grey color representing CTRL group, blue color representing HS320 group, red color representing HS350 group. Results are expressed as geometric mean fluorescence intensity (GMFI). DHE—dihydroethidium; CTRL—control; HS—high salt; One-way ANOVA; *<sup>,+</sup> significance level <span class="html-italic">p</span> &lt; 0.05 (* compared to HS group before AngII; <sup>+</sup> compared to control group).</p>
Full article ">Figure 4
<p>Changes in CAMs’ expression: VCAM-1 (<b>A</b>), ICAM-1 (<b>B</b>), Endoglin (<b>C</b>), and E-selectin (<b>D</b>) in HAECs following high-salt treatment accompanied by AngII exposure. Grey color representing CTRL group, blue color representing HS320 group, red color represent-ing HS350 group. Results are expressed as geometric mean fluorescence intensity (GMFI). CAMs—cell adhesion molecules; HAECs—human aortic endothelial cells; VCAM-1—vascular cell adhesion molecule 1; ICAM-1—intracellular adhesion molecule 1; HS—high salt; CTRL—control; AngII—angiotensin II; One-way ANOVA; *<sup>,+</sup> significance level <span class="html-italic">p</span> &lt; 0.05 (* compared to HS group before AngII; <sup>+</sup> compared to control group).</p>
Full article ">
47 pages, 3709 KiB  
Review
Oxidative Stress in Kidney Injury and Hypertension
by Willaim J. Arendshorst, Aleksandr E. Vendrov, Nitin Kumar, Santhi K. Ganesh and Nageswara R. Madamanchi
Antioxidants 2024, 13(12), 1454; https://doi.org/10.3390/antiox13121454 - 27 Nov 2024
Viewed by 584
Abstract
Hypertension (HTN) is a major contributor to kidney damage, leading to conditions such as nephrosclerosis and hypertensive nephropathy, significant causes of chronic kidney disease (CKD) and end-stage renal disease (ESRD). HTN is also a risk factor for stroke and coronary heart disease. Oxidative [...] Read more.
Hypertension (HTN) is a major contributor to kidney damage, leading to conditions such as nephrosclerosis and hypertensive nephropathy, significant causes of chronic kidney disease (CKD) and end-stage renal disease (ESRD). HTN is also a risk factor for stroke and coronary heart disease. Oxidative stress, inflammation, and activation of the renin–angiotensin–aldosterone system (RAAS) play critical roles in causing kidney injury in HTN. Genetic and environmental factors influence the susceptibility to hypertensive renal damage, with African American populations having a higher tendency due to genetic variants. Managing blood pressure (BP) effectively with treatments targeting RAAS activation, oxidative stress, and inflammation is crucial in preventing renal damage and the progression of HTN-related CKD and ESRD. Interactions between genetic and environmental factors impacting kidney function abnormalities are central to HTN development. Animal studies indicate that genetic factors significantly influence BP regulation. Anti-natriuretic mechanisms can reset the pressure–natriuresis relationship, requiring a higher BP to excrete sodium matched to intake. Activation of intrarenal angiotensin II receptors contributes to sodium retention and high BP. In HTN, the gut microbiome can affect BP by influencing energy metabolism and inflammatory pathways. Animal models, such as the spontaneously hypertensive rat and the chronic angiotensin II infusion model, mirror human essential hypertension and highlight the significance of the kidney in HTN pathogenesis. Overproduction of reactive oxygen species (ROS) plays a crucial role in the development and progression of HTN, impacting renal function and BP regulation. Targeting specific NADPH oxidase (NOX) isoforms to inhibit ROS production and enhance antioxidant mechanisms may improve renal structure and function while lowering blood pressure. Therapies like SGLT2 inhibitors and mineralocorticoid receptor antagonists have shown promise in reducing oxidative stress, inflammation, and RAAS activity, offering renal and antihypertensive protection in managing HTN and CKD. This review emphasizes the critical role of NOX in the development and progression of HTN, focusing on its impact on renal function and BP regulation. Effective BP management and targeting oxidative stress, inflammation, and RAAS activation, is crucial in preventing renal damage and the progression of HTN-related CKD and ESRD. Full article
(This article belongs to the Special Issue NADPH Oxidases in Health and Aging—2nd Edition)
Show Figures

Figure 1

Figure 1
<p><b>An intricate network of pathways leads to renal nephrosclerosis in hypertension.</b> Hypertension and RAAS (renin–angiotensin–aldosterone system) activation initiate a cascade of events, including NOX activation and ROS generation, which induce endothelial dysfunction, inflammation, and epithelial-to-mesenchymal transition (EMT). The resulting podocyte injury and apoptosis contribute to the denudation of the glomerular basement membrane. Concurrently, endothelial-to-mesenchymal transition (EndoMT), vascular smooth muscle cell (VSMC) proliferation, and myofibroblast activation occur, leading to hyalinosis and narrowing of the afferent arteriole, the collapse of capillary loops, and retraction of the glomerular tuft. Epithelial cell damage and EMT lead to tubular dilation, atrophy, inflammation, and fibrosis, ultimately resulting in tubulointerstitial fibrosis and the filling of Bowman’s space with collagen.</p>
Full article ">Figure 2
<p><b>Oxidative stress is central to the pathophysiological pathways leading to hypertension.</b> Unhealthy dietary habits and antibiotic use disrupt the gut microbiome, leading to an imbalance that promotes the release of pro-inflammatory cytokines and metabolites and the activation of immune cells. The presence of inflammatory mediators in conjunction with other comorbidities results in reduced nitric oxide (NO) bioavailability and impaired activities of SOD and catalase, inducing mitochondrial dysfunction and increased ROS generation. This, in turn, triggers endoplasmic reticulum (ER) stress and the unfolded protein response (UPR). Critical signaling pathways like TGFβ/SMAD2/3, NFκB, and RAAS) are activated, further worsening oxidative stress by upregulating NOX enzyme levels. Oxidative stress is a pivotal downstream event in the pathophysiological cascade, causing cellular dysfunction, damage, and apoptosis. In the kidney, oxidative stress induces renal inflammation, fibrosis, endothelial dysfunction, and VSMC proliferation. These processes result in renal vasoconstriction, vascular hypertrophy, remodeling, and increased sodium and water retention, leading to elevated systemic vascular resistance and blood pressure, ultimately causing hypertension.</p>
Full article ">Figure 3
<p><b>Mechanisms affecting renal function and pressure–natriuresis relation in the development of hypertension.</b> The pressure–natriuresis relationship describes the kidney’s ability to excrete Na<sup>+</sup> in response to changes in BP. Increased SNS and RAAS activity, renal oxidative stress, and inflammation at normotensive BP result in impaired pressure–natriuresis, characterized by reduced Na<sup>+</sup> excretion. That leads to elevated BP and a “reset” of the pressure–natriuresis curve, normalizing Na<sup>+</sup> excretion to match sodium intake levels. This normalization is characterized by improved Na<sup>+</sup> filtration capacity and reduced Na<sup>+</sup> retention. Up arrows indicate an increased effect, while down arrows signify a decreased effect.</p>
Full article ">Figure 4
<p><b>NADPH oxidases expression in the renal cells.</b> NADPH oxidases (NOX) and mitochondria are the primary sources of ROS in renal and vascular cells, including VSMC, podocytes, mesangial cells, and tubular epithelial cells.</p>
Full article ">Figure 5
<p><b>Animal models of hypertension.</b> In the Dahl salt-sensitive rat model, a high salt diet leads to renal inflammation, increased ROS levels, activation of the SNS and RAAS, decreased GFR, and changes in Na<sup>+</sup> and water excretion. The chronic infusion of Ang II leads to similar pathophysiological changes, including increased ROS levels, RAAS activation, impaired Na<sup>+</sup> excretion, reduced GFR, endothelial dysfunction, and heightened renal vascular reactivity and resistance. In the spontaneously hypertensive rat model, HTN develops through genetic predisposition when the animals are fed a normal salt diet. Pathophysiological changes involve renal inflammation, oxidative stress, RAAS activation, Na<sup>+</sup> retention, and impaired pressure–natriuresis. Preventive measures across all models include RAAS inhibition (ACEi/ARB), antioxidants, and dietary modifications such as a low or normal salt diet.</p>
Full article ">Figure 6
<p><b>Renal and systemic effects of proximal tubular SGLT2 (sodium–glucose cotransporter-2) inhibition.</b> SGLT2 inhibitors lead to glucosuria and promote renal natriuresis by enhancing mitochondrial function and reducing oxidative stress. This improvement helps to enhance glomerular function and decrease tubulointerstitial fibrosis. On a systemic level, SGLT2 inhibition results in lower plasma glucose levels, reduced renal and systemic inflammation, decreased vasoconstriction and vascular injury, Na<sup>+</sup> retention, and BP.</p>
Full article ">
18 pages, 1302 KiB  
Article
Potentiation of NMDA Receptors by AT1 Angiotensin Receptor Activation in Layer V Pyramidal Neurons of the Rat Prefrontal Cortex
by Adrienn Hanuska, Polett Ribiczey, Erzsébet Kató, Zsolt Tamás Papp, Zoltán V. Varga, Zoltán Giricz, Zsuzsanna E. Tóth, Katalin Könczöl, Ákos Zsembery, Tibor Zelles, Laszlo G. Harsing and László Köles
Int. J. Mol. Sci. 2024, 25(23), 12644; https://doi.org/10.3390/ijms252312644 - 25 Nov 2024
Viewed by 368
Abstract
NMDA receptors in the prefrontal cortex (PFC) play a crucial role in cognitive functions. Previous research has indicated that angiotensin II (Ang II) affects learning and memory. This study aimed to examine how Ang II impacts NMDA receptor activity in layer V pyramidal [...] Read more.
NMDA receptors in the prefrontal cortex (PFC) play a crucial role in cognitive functions. Previous research has indicated that angiotensin II (Ang II) affects learning and memory. This study aimed to examine how Ang II impacts NMDA receptor activity in layer V pyramidal cells of the rat PFC. Whole-cell patch-clamp experiments were performed in pyramidal cells in brain slices of 9–12-day-old rats. NMDA (30 μM) induced inward currents. Ang II (0.001–1 µM) significantly enhanced NMDA currents in about 40% of pyramidal cells. This enhancement was reversed by the AT1 antagonist eprosartan (1 µM), but not by the AT2 receptor antagonist PD 123319 (5 μM). When pyramidal neurons were synaptically isolated, the increase in NMDA currents due to Ang II was eliminated. Additionally, the dopamine D1 receptor antagonist SCH 23390 (10 μM) reversed the Ang II-induced enhancement, whereas the D2 receptor antagonist sulpiride (20 μM) had no effect. The potentiation of NMDA currents in a subpopulation of layer V pyramidal neurons by Ang II, involving AT1 receptor activation and dopaminergic signaling, may serve as an underlying mechanism for the effects of the renin–angiotensin system (RAS) elements on neuronal functions. Full article
(This article belongs to the Special Issue The Role of Neurons in Human Health and Disease—3rd Edition)
Show Figures

Figure 1

Figure 1
<p>NMDA-evoked inward currents in layer V pyramidal neurons of the rat prefrontal cortex. Whole-cell patch-clamp measurements at a holding potential of −70 mV. Correlation of concentration–response amplitudes for NMDA-induced currents. Each data point corresponds to measurements taken from <span class="html-italic">n</span> cells for each NMDA concentration (10 µM, <span class="html-italic">n</span> = 3; 30 µM, <span class="html-italic">n</span> = 6; 100 µM, <span class="html-italic">n</span> = 7; 300 µM, <span class="html-italic">n</span> = 6).</p>
Full article ">Figure 2
<p>30 µM NMDA-induced inward currents in layer V pyramidal neurons of the rat PFC. (<b>A</b>) Diagram of the experimental patch-clamp protocol. After the whole-cell configuration was established, 200 µm thick mPFC slices were superfused with aCSF for 10 min to achieve diffusion balance between the patch pipette and the cell interior. Then, 30 μM NMDA was applied three times for 1.5 min (T<sub>1</sub>, T<sub>2</sub>, T<sub>3</sub>), separated by superfusion periods of 10 min with drug-free aCSF. The membrane currents were measured using the amplifier in voltage-clamp mode at a holding potential of −70 mV. The amplitude of the NMDA-induced ion currents was quantified. (<b>B</b>) Representative tracing of the current response to NMDA after three applications of 30 μM NMDA (T<sub>1</sub>, T<sub>2</sub>, T<sub>3</sub>).</p>
Full article ">Figure 3
<p>Effect of 1 nM–1 µM Ang II on NMDA-induced inward currents in layer V pyramidal neurons of the rat prefrontal cortex. Whole-cell patch-clamp measurements were conducted at a holding potential of −70 mV. A total of 30 µM of NMDA was applied three times for 1.5 min (T<sub>1</sub>, T<sub>2</sub>, T<sub>3</sub>) with a 10 min interval between applications. Under these conditions, current responses were consistent at T<sub>2</sub> and T<sub>3</sub>. Ang II at concentrations of 1 nM–1 µM was applied for 5 min before and during T<sub>3</sub>. (<b>A</b>) Mean ± SEM of <span class="html-italic">n</span> experiments, showing the effects of 0 µM (control) (<span class="html-italic">n</span> = 19), 1 nM (<span class="html-italic">n</span> = 11/28), 10 nM (<span class="html-italic">n</span> = 9/22), and 1 µM (<span class="html-italic">n</span> = 21/47) Ang II on NMDA currents with respect to the response measured at T2. Green bars represent the normalized NMDA-induced current responses (%) in the subpopulation of mPFC layer V pyramidal cells in which Ang II potentiated ion currents. * <span class="html-italic">p</span> &lt; 0.05, a significant difference from the normalized current responses to NMDA under control conditions. (<b>B</b>) Representative tracing of a current response to 30 μM NMDA after T<sub>2</sub> and T<sub>3</sub>, and in the presence of 1 µM Ang II for 5 min before and during T<sub>3</sub>.</p>
Full article ">Figure 4
<p>Role of AT<sub>1</sub> and AT<sub>2</sub> receptors in the potentiation of NMDA receptor function by Ang II and protein/mRNA expression of AT<sub>1</sub> receptors (AT1R) in the rat mPFC. (<b>A</b>) NMDA-induced current responses in mPFC layer V pyramidal cells were detected using whole-cell patch-clamp measurements at a holding potential of −70 mV. A 30 µM concentration of NMDA was administered three times for 1.5 min each (T<sub>1</sub>, T<sub>2</sub>, T<sub>3</sub>) with a 10 min interval between applications. A 1 µM concentration of Ang II was applied for 5 min before and during T<sub>3</sub>. In separate experiments, aCSF contained either 1 µM eprosartan (AT<sub>1</sub> antagonist) or 5 µM PD 123319 (AT<sub>2</sub> antagonist) throughout the entire measurement period. The data are presented as the mean ± SEM of <span class="html-italic">n</span> experiments: effects of 1 µM Ang II (<span class="html-italic">n</span> = 21/47), 1 µM Ang II + 1 µM eprosartan (<span class="html-italic">n</span> = 8), and 1 µM Ang II + 5 µM PD 123319 (<span class="html-italic">n</span> = 5/11) on NMDA currents at T<sub>3</sub>, normalized with respect to the response measured at T<sub>2</sub>. Green bars indicate normalized NMDA-induced current responses (T<sub>3</sub>/T<sub>2</sub> %) in cells where Ang II potentiated NMDA receptor-mediated ion currents. If potentiation cannot be observed, all cells in the group are represented. * <span class="html-italic">p</span> &lt; 0.05 indicates a significant difference from the 1 µM Ang II potentiation group. (<b>B</b>) Immunohistochemical detection of AT<sub>1</sub> receptor protein expression (<b>left</b> panel) was conducted using the MBS151548 anti-AT1R rabbit polyclonal antibody, and fluorescent in situ hybridization analysis of AT1R mRNA expression (<b>right</b> panel) was performed in the mPFC of 10-day-old Wistar rats. For immunohistochemical detection, the MBS151548 anti-AT1R rabbit polyclonal antibody preincubated with the MBS152017 AT1R blocking peptide served as the negative control (<b>left</b> panel, square below). Blue indicates cells counterstained with 4′,6-diamidino-2-phenylindole (DAPI), green indicates cells stained with anti-AT1R rabbit polyclonal antibody (left panel), and cells expressing AT1R mRNA (<b>right</b> panel). L1, L2/3, and L5/6 denote the layers of the mPFC.</p>
Full article ">Figure 5
<p>The effect of synaptic isolation and the role of D1 and D2 dopaminergic receptors in Ang II-induced NMDA receptor potentiation. NMDA-induced current responses in mPFC layer V pyramidal cells were detected using whole-cell patch-clamp measurements at a holding potential of −70 mV. A total of 30 µM of NMDA was administered three times for 1.5 min (T<sub>1</sub>, T<sub>2</sub>, T<sub>3</sub>) with a 10 min interval between applications. A total of 1 µM of Ang II was applied for 5 min before and during T<sub>3</sub>. To test the effect of synaptic isolation, 0.5 µM tetrodotoxin (TTX) was added to the aCSF, or Ca<sup>2+</sup>-free aCSF was used throughout the entire experiment. To detect D1 receptor and D2 receptor involvement in Ang II-induced potentiation of the NMDA receptor, 10 µM SCH-23390 (D1 receptor antagonist) or 20 µM sulpiride (D2 receptor antagonist) was added to the aCSF throughout the measurements. Mean ± SEM of <span class="html-italic">n</span> experiments: effects of 1 µM Ang II (<span class="html-italic">n</span> = 21/47), 1 µM Ang II + 0.5 µM TTX (<span class="html-italic">n</span> = 8), 1 µM Ang II + Ca<sup>2+</sup>-free aCSF (<span class="html-italic">n</span> = 15), 1 µM Ang II + 10 µM SCH-23390 (<span class="html-italic">n</span> = 8), and 1 µM Ang II + 20 µM sulpiride (<span class="html-italic">n</span> = 8/20) on NMDA currents at T<sub>3</sub> normalized with respect to the response measured in T<sub>2</sub>. Green bars indicate normalized NMDA-induced current responses (T<sub>3</sub>/T<sub>2</sub> %) in cells where Ang II potentiated NMDA receptor-mediated ion currents. If potentiation cannot be observed, all cells in the group are represented. * <span class="html-italic">p</span> &lt; 0.05 indicates a significant difference from the 1 µM Ang II potentiation group.</p>
Full article ">
20 pages, 982 KiB  
Review
Correlation Between Antihypertensive Drugs and Survival Among Patients with Pancreatic Ductal Adenocarcinoma
by Natalia Kluz, Leszek Kraj, Paulina Chmiel, Adam M. Przybyłkowski, Lucjan Wyrwicz, Rafał Stec and Łukasz Szymański
Cancers 2024, 16(23), 3945; https://doi.org/10.3390/cancers16233945 - 25 Nov 2024
Viewed by 529
Abstract
There is a growing prevalence of pancreatic cancer, accompanied by accelerated disease progression and diminished survival rates. Radical resection with clear margins remains the sole viable option for achieving a long-term cure in patients. In cases of advanced, unresectable, and metastatic disease, chemotherapy [...] Read more.
There is a growing prevalence of pancreatic cancer, accompanied by accelerated disease progression and diminished survival rates. Radical resection with clear margins remains the sole viable option for achieving a long-term cure in patients. In cases of advanced, unresectable, and metastatic disease, chemotherapy based on leucovorin, 5-fluorouracil, irinotecan, oxaliplatin, gemcitabine, or nab-paclitaxel represents the cornerstone of the treatment. Considering the limited treatment options available following initial therapy, the strategy of repurposing commonly prescribed drugs such as antihypertensives into anti-cancer therapies in palliative treatment represents a promising avenue for enhancing survival in patients with pancreatic ductal adenocarcinoma. The repurposing of existing drugs is typically a more cost-effective and expedient strategy than the development of new ones. The potential for antihypertensive drugs to be employed as adjunctive therapies could facilitate a more comprehensive treatment approach by targeting multiple pathways involved in cancer progression and acquired resistance to treatment. Antihypertensive medications, particularly those belonging to the pharmacological classes of angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers, and calcium channel blockers, are commonly prescribed and have well-established safety profiles, particularly among patients with pancreatic cancer who are affected by multiple comorbidities. Therefore, we emphasize the preclinical and clinical evidence supporting the use of antihypertensive agents in the treatment of pancreatic cancer, emphasizing their beneficial chemosensitizing effects. Full article
(This article belongs to the Special Issue Advanced Research in Pancreatic Ductal Adenocarcinoma)
Show Figures

Figure 1

Figure 1
<p>Antihypertensive drugs in PDAC: antitumoral mechanisms. In this figure, we summarize potential mechanisms through which antihypertensive drugs may aid chemotherapy through different cellular effects. Ag II, angiotensin II; HA, hyaluronic acid; ACEI, angiotensin-converting enzyme inhibitors; ARB, angiotensin I receptor blocker; CCB, calcium channel blocker; β2AR, β2-adrenergic receptor; BBs, beta-blockers. Created in BioRender.com (accessed on 20 November 2024).</p>
Full article ">
Back to TopTop